header logo image


Page 274«..1020..273274275276..280290..»

Initiation of First UniCAR Cellular Immunotherapy Study in Advanced Solid Tumors – BioSpace

December 4th, 2020 12:16 am

DRESDEN, Germany, Dec. 3, 2020 /PRNewswire/ -- GEMoaB, a biopharmaceutical company focused on the development of next-generation immunotherapies for hard-to-treat cancers, today announced that it has apheresed the first patient in a Phase IA study with UniCAR-T-PSMA, the lead solid tumor product candidate from its proprietary UniCAR cellular immunotherapy platform. UniCAR-T-PSMA is investigated in late-stage, relapsed/refractory solid tumors expressing the PSMA antigen. The UniCAR platform has been designed to ensure excellent control over the universal CAR-T effector cell through a rapidly switchable on/off capability. This is combined with high flexibility to effectively target tumor antigens of choice by re-directing and activating UniCAR-T cells through soluble adapters termed Targeting Modules (TMs).

"The unique ability to rapidly switch on and off the UniCAR-T effector cells and thereby tightly control their activity as we have now clinically validated in our ongoing UniCAR-T-CD123 study in AML may help to overcome many of the limitations that conventional CAR-T therapies face when targeting less differentially expressed antigens, especially in solid tumors", said Prof. Dr. Gerhard Ehninger, GEMoaB's co-founder and Chief Medical Officer. "Our first UniCAR clinical study in solid tumors is of utmost importance for GEMoaB. We believe that the PSMA antigen is a great initial target as it is not only expressed on the tumor surface but also on the tumor neo-vasculature, allowing for a double attack of the malignant cells by UniCAR-T cells."

The Phase IA study includes patients with late-stage PSMA-positive relapsed/refractory solid tumors such as Castrate Resistant Prostate Cancer (CRPC), Non-small Cell Lung Cancer (NSCLC) or Triple Negative Breast Cancer (TNBC). It will examine the feasibility, safety and potential efficacy of the combined application of a single dose of UniCAR-T and the continuous infusion of the PSMA-specific TMpPSMA.

According to Prof. Dr. Ralf Bargou, Head of Comprehensive Cancer Center Mainfranken at the University Hospital Wrzburg and coordinating investigator of this trial, the study could be an important step in the ongoing intensive research efforts to establish cellular immunotherapies as a key therapeutic pillar to improve patient outcomes in hard-to-treat solid tumor cancers. "At our National Cancer Center in Wrzburg we are focusing a significant amount of our ongoing research and clinical efforts on developing breakthrough immunologic treatments of solid tumors together with our partners", said Prof. Bargou. "PSMA is a very promising target expressed in multiple late-stage cancers that do not sufficiently benefit from currently existing therapies and the UniCAR platform provides many features to finally obtain meaningful safety and efficacy results for this innovative treatment modality. We are very much looking forward working closely with the GEMoaB team on this important study."

About the UniCAR-T-PSMA Study

This first-in-human phase I study is an open-label, non-randomized, dose-finding study designed to evaluate the safety and activity of UniCAR-T-PSMA in up to 16 patients with advanced relapsed/refractory, PSMA-positive solid tumors such as CRPC, NSCLC or TNBC. Its purpose is to determine the maximum tolerated dose (MTD), dose limiting toxicities (DLT) as well as the recommended Phase II dose for the combined application of a single dose of UniCAR-T and the continuous infusion of TMpPSMA over 25 days. The study will also investigate response rates, persistence of UniCAR-T cells over time as well as the ability to rapidly switch UniCAR-T cells on and off in case of side effects through stopping the TM infusion. The study will take place at selected Phase I and CAR-T experienced University centers in Germany. It is supported by a grant from the European Regional Development Fund (ERDF) provided through Saxony's Development Bank (SAB). To learn more about the trial, please visit clinicaltrials.gov.

About UniCAR

GEMoaB is developing a rapidly switchable universal CAR-T platform, UniCAR, to improve the therapeutic window and increase efficacy and safety of CAR-T cell therapies in challenging cancers, including acute leukemias and solid tumors. Conventional CAR-T cells depend on the presence and direct binding of cancer antigens for activation and proliferation. An inherent key feature of the UniCAR platform is a rapidly switchable on/off mechanism (less than 4 hours after interruption of TM supply) enabled by the short pharmacokinetic half-life and fast internalization of soluble adaptors termed TMs. These TMs provide the antigen-specificity to activate UniCAR gene-modified T-cells (UniCAR-T) and consist of a highly flexible antigen binding moiety, linked to a small peptide motif recognized by UniCAR-T.

About GEMoaB

GEMoaB is a privately-owned, clinical-stage biopharmaceutical company that is aiming to become a fully integrated biopharmaceutical company. By advancing its proprietary UniCAR, RevCAR and ATAC platforms, the company will discover, develop, manufacture and commercialize next-generation immunotherapies for the treatment of cancer patients with a high unmet medical need.

GEMoaB has a broad pipeline of product candidates in pre-clinical and clinical development for the treatment of hematological malignancies as well as solid tumors. Its clinical stage assets GEM333, an Affinity-Tailored Adaptor for T-Cells (ATAC) with binding specificity to CD33 in relapsed/refractory AML, and GEM3PSCA, an ATAC with binding specificity to PSCA for the treatment of castrate-resistant metastatic prostate cancer and other PSCA expressing late stage solid tumors, are currently investigated in Phase I studies and globally partnered with Bristol-Myers Squibb. A Phase IA dose-finding study of the first UniCAR asset in hematological malignancies, UniCAR-T-CD123 for treatment of relapsed/refractory AML, is currently recruiting patients.

Manufacturing expertise, capability and capacity are key for developing cellular immunotherapies for cancer patients. GEMoaB has established a preferred partnership with its sister company Cellex in Cologne, a world leader in manufacturing hematopoietic blood stem cell products and a leading European CMO for CAR-T cells, co-operating in that area with several large biotech companies.

More information can be found at http://www.gemoab.com.

For further information please contact

Jana Fiebigerj.fiebiger@gemoab.com; Tel.: +49 351 4466-45012

Investor Contact

Michael Pehlm.pehl@gemoab.com; Tel.: +49 351 4466-45030

Forward-looking Statements This announcement includes forward-looking statements that involve risks, uncertainties and other factors, many of which are outside of our control, that could cause actual results to differ materially from the results and matters discussed in the forward looking statements. Forward looking statements include statements concerning our plans, goals, future events and or other information that is not historical information. The Company does not assume any liability whatsoever for forward-looking statements. The Company assumes that potential partners will perform and rely on their own independent analyses as the case may be. The Company will be under no obligation to update the Information.

Logo- https://mma.prnewswire.com/media/947610/GEMoaB_Logo.jpg

View original content:http://www.prnewswire.com/news-releases/initiation-of-first-unicar-cellular-immunotherapy-study-in-advanced-solid-tumors-301185515.html

SOURCE GEMoaB

See the rest here:
Initiation of First UniCAR Cellular Immunotherapy Study in Advanced Solid Tumors - BioSpace

Read More...

Transcriptome and regulatory maps of decidua-derived stromal cells inform gene discovery in preterm birth – Science Advances

December 4th, 2020 12:16 am

Abstract

While a genetic component of preterm birth (PTB) has long been recognized and recently mapped by genome-wide association studies (GWASs), the molecular determinants underlying PTB remain elusive. This stems in part from an incomplete availability of functional genomic annotations in human cell types relevant to pregnancy and PTB. We generated transcriptome (RNA-seq), epigenome (ChIP-seq of H3K27ac, H3K4me1, and H3K4me3 histone modifications), open chromatin (ATAC-seq), and chromatin interaction (promoter capture Hi-C) annotations of cultured primary decidua-derived mesenchymal stromal/stem cells and in vitro differentiated decidual stromal cells and developed a computational framework to integrate these functional annotations with results from a GWAS of gestational duration in 56,384 women. Using these resources, we uncovered additional loci associated with gestational duration and target genes of associated loci. Our strategy illustrates how functional annotations in pregnancy-relevant cell types aid in the experimental follow-up of GWAS for PTB and, likely, other pregnancy-related conditions.

Spontaneous preterm birth (PTB), defined as spontaneous labor and birth before 37 weeks of gestation, is associated with considerable infant mortality and morbidity, as well as long-term health consequences into adulthood (1). A genetic component to PTB has long been recognized, but the significant role of environmental factors and the etiologic heterogeneity of birth before 37 weeks (24) have made it challenging to discover genetic associations and causal genes. For example, recent genome-wide association studies (GWASs) of gestational duration in 43,568 women (3331 with a preterm delivery) (5) and in 84,689 infants (4775 born preterm) (6) reported six and one genome-wide significant associations, respectively, with gestational duration considered as a continuous variable. Three loci were also associated with PTB (defined as a categorical variable of birth) in the maternal GWAS (5), but no loci were associated with PTB in the infant GWAS (6). These studies highlight the challenges of such complex and multifactorial phenotypes and the need for additional approaches to facilitate discovery of genes contributing to gestational duration and PTB.

Integrating GWAS that results with genomic and epigenomic annotations is a promising approach for assigning function to variants discovered by GWAS, as well as for identifying additional associations that do not reach stringent genome-wide significance threshold (7, 8). While large consortia [e.g., ENCODE (Encyclopedia of DNA Elements) (9), GTEx (Genotype-Tissue Expression Project) (10), and Roadmap Epigenomics (11)] have generated annotations of putative functional elements and genetic variants for many human cell types and tissues, there is a remarkable absence in these databases for the cell types and tissues that are relevant to pregnancy in general and to PTB in particular. Because the regulation of transcription has strong cell typespecific components and because annotations in disease-relevant tissues or cells tend to be most enriched among GWAS signals for those specific diseases (10, 12), follow-up studies of GWASs of pregnancy-associated conditions have been disadvantaged compared to most other complex diseases due to the paucity of functional annotations in cells relevant to pregnancy. To fill this gap in knowledge, we characterized the transcriptional and chromatin landscapes of cultured mesenchymal stromal/stem cells (MSCs) collected from human placental membranes and decidualized MSCs, also known as decidual stromal cells (DSCs). These cells play critical roles in promoting successful pregnancy, interfacing with fetal cells throughout pregnancy, and the timing of birth (13, 14). We then built a computational framework that integrated these decidua-derived stromal cell annotations with the results of a large GWAS of gestational duration to facilitate discovery of PTB genes.

This integrated analysis revealed a significant enrichment of heritability estimates for gestational duration in decidua-derived stromal cell genomic regions marked by open chromatin or histone marks. Leveraging those functional annotations in a Bayesian statistical framework, we discovered additional loci associated with gestational duration and improved fine mapping in regions associated with gestational duration. Last, using promoter capture Hi-C (pcHi-C), we linked functionally annotated gestational age-associated variants to their putative target genes. More generally, these functional annotations should prove a valuable resource for studying other pregnancy-related conditions, such as preeclampsia and recurrent miscarriage, as well as conditions associated with endometrial dysfunction, such as endometriosis and infertility.

Decidualization is the process of transformation of endometrial MSCs into DSCs that is induced by progesterone production that begins during the luteal phase of the menstrual cycle and then increases throughout pregnancy when successful implantation occurs [reviewed in (15)]. Using progesterone and estrogen or cyclic adenosine 5-monophosphate (cAMP) to induce decidualization of MSCs in culture has been used in cells derived from endometrial biopsies in nonpregnant women to characterize their transcriptomes and epigenomes and to identify genes and molecular pathways involved in this process (1621).

Because obtaining endometrial cells in nonpregnant women through biopsies requires an invasive procedure that carries some risk and MSCs can also be obtained from human placentas (2224), we isolated these cells from the decidua parietalis of three women who had delivered at term and established one primary MSC line from each to model the process of decidualization (see Materials and Methods). Briefly, cells were treated with medroxyprogesterone acetate (MPA) and cAMP for 48 hours, and a paired set of untreated samples was cultured in parallel for 48 hours. Three replicates of treated/untreated sets of each cell line were studied to assess experimental variability in the two conditions. Each of the 18 samples (3 individual lines 3 replicates 2 conditions) were assayed to generate transcriptome [RNA sequencing (RNA-seq)], open chromatin [assay for transposase-accessible chromatin sequencing (ATAC-seq)], and histone modification [chromatin immunoprecipitation sequencing (ChIP-seq)] maps. A summary of those data is shown in table S1, and a representative example of the full set of annotations for one primary cell line is shown in Fig. 1. The number of reads generated for each sample in each condition and other descriptive data are provided in data file S1.

Each histone modification and RNA-seq track shows read counts per base pair for each experiment. The pcHi-C signal track shows the number of reads per MboI restriction fragment. Arcs in the pcHi-C interactions track show significant interactions between the promoter of the PRL gene and putative distal regulatory elements identified with pcHi-C. Pooled data (three replicates) for one cell line are shown for untreated cells (MSCs, in green) and decidualized cells (DSCs, in purple). pcHi-C data were generated in a fourth cell line that was decidualized.

Analysis of the RNA-seq data using DESeq2 (25) revealed 1135 differentially expressed genes after decidualization (table S1). Genes with decreased expression after 48 hours of treatment were highly enriched for cell cycle genes (data file S2), consistent with observations from endometrial biopsies from nonpregnant women that decidualization is associated with cell cycle arrest (19, 26). Genes with increased expression after treatment were enriched for insulin-related terms, also consistent with previous results from endometrial biopsies (26), and for glucose metabolism (18).

To identify putative regulatory elements in MSCs and DSCs, we assayed H3K27ac, H3K4me1, and H3K4me3 histone modifications, which are markers of active enhancers, poised enhancers, and active promoters, respectively [reviewed in (27)]. We also used ATAC-seq to identify open chromatin regions to complement ChIP-seq data. To identify regulatory regions that might be altered in response to, and potentially regulate decidualization, we compared read counts of ATAC-seq and ChIP-seq peaks in untreated and decidualized cells, revealing tens of thousands of regions that differed between untreated and treated samples (table S1). Most of the differential peaks were marked with H3K27Ac and H3K4me1, indicating that the epigenetic changes underlying alterations in gene expression during decidualization predominantly occur in distant regulatory elements, such as enhancers.

We observed a moderate degree of overlap between the differential peaks across ATAC-seq and ChIP-seq data, with the two enhancer marks, H3K27ac and H3K4me1, showing the most overlap (Fig. 2A). In addition, putative regulatory regions that showed chromatin changes in response to decidualization were associated with genes whose expression also changed in response to decidualization (Fig. 2B). Regulatory regions with increased read counts clustered around genes that were more highly expressed after decidualization, indicating increased chromatin accessibility or activation of enhancers of those genes. Conversely, genes that were more lowly expressed after decidualization were enriched for enhancers that became less accessible or active. These observations indicate that the differential peaks of open chromatin and histone marks observed after decidualization correspond to regulatory elements that become more or less active, resulting in correlated gene expression changes of the nearby genes.

(A) Plot showing the overlap between the different histone modifications and ATAC-seq maps (intersection between annotations). Peaks were assigned to 100-bp bins to avoid ambiguity in overlap due to different peak borders. Black circles indicate overlap with other annotations; light gray circles indicate that the annotation does not overlap others. (B) Each data point shows the ratio between the number of increased/decreased differential peaks nearby genes that increase expression after decidualization (blue, positive log ratios; upper half of the figure) or decrease expression after decidualization (orange, negative log ratios; lower half of the figure). Genes that were more highly expressed in decidualized cells were flanked by a higher number of ChIP-seq and ATAC-seq peaks that displayed increased read counts in decidualized samples compared to peaks that displayed decreased read counts (top inset). Genes that were down-regulated in decidualized cells showed the opposite trend (bottom inset). All enrichments: P < 1025. (C) DNA binding motifs of transcription factors relevant in decidualization are enriched in peaks that change following decidualization treatment. Motifs are color-coded by similarity. (D) Colocalization of PGR, FOSL2, FOXO1, GATA2, and NR2F2 with ATAC-seq and ChIP-seq peaks. Transcription factor binding sites co-occur with ATAC-seq and ChIP-seq peaks in both untreated (green) and decidualized (purple) cells more often than with random peaks. Enrichment of the co-occurrences of PGR, FOXO1, GATA2, and NR2F2 are higher when co-occurring with peaks that have increased read counts (navy blue) and lower with peaks that have decreased read counts (orange) in decidualized compared to untreated cells. Enrichment of co-occurrences with peak sets was calculated as the fold difference between the number of transcription factor peaks overlapping with ATAC-seq/ChIP-seq peaks and with a random set of peaks (see Materials and Methods).

Previous work identified transcription factors that play critical roles in decidualized stromal cells (2832). Several of the DNA binding motifs that were enriched in peaks with increased or decreased read counts in our data correspond to transcription factors previously implicated in decidualization (Fig. 2C), such as CAAT-enhancer binding protein (CEBP) (33), progesterone receptor (PGR) (28) that shares the same motif with androgen response element, and glucocorticoid receptor, FOSL2 (Fos-related antigen 2) (28), that shares the same motif with Fra1 (Fos-related antigen 1), Atf3 (Activating transcription factor 3), and BATF (basic leucine zipper ATF-like transcription factor), and TEA (transcriptional enhancer factor) domain transcription factors (21, 34). Whereas CEBP and PGR were exclusively enriched in peaks with increased read counts in decidualized cells, the FOSL2 motif was present in peaks that both changed positively and negatively in decidualized cells.

To better understand the role of these transcription factors in decidualization, we obtained publicly available ChIP-seq data for PGR (28) and FOSL2 (28) from endometrial biopsies and analyzed the colocalization of their binding locations with the putative regulatory elements identified by ATAC-seq and ChIP-seq identified in our study (Fig. 2B). We additionally analyzed FOXO1 (Forkhead box O1) (29), NR2F2 (nuclear receptor subfamily 2 group F member 2) (30), and GATA2 (GATA binding protein 2) (31) ChIP-seq data because these transcription factors have also been implicated in decidualization (2931). With the exception of FOSL2, the colocalization enrichments of PGR, FOXO1, GATA2, and NR2F2 with ATAC-seq and ChIP-seq peaks were higher (9 to 16 folds) among peaks that were increased in decidualized cells (more open chromatin or increased histone modification levels) compared to all peaks (7.5 to 12.8 folds) and to peaks that decreased in decidualized cells (2 to 5 folds). This observation supports the notion that these transcription factors are involved in regulation of decidualization (2831, 35). Although FOSL2 has been reported as a positive coregulator of PGR (28), the presence of FOSL2 motifs in peaks that both increased and decreased in decidualized cells (Fig. 2C) and the lack of difference in the colocalization enrichment between these two sets of peaks (Fig. 2D) suggests that FOSL2 may have a dual role in decidualization.

Together, our results support a model of decidualization that involves changes in the regulatory landscape during the differentiation of MSCs into DSCs, including alterations in chromatin accessibility and in the activation levels of distant regulatory elements, accompanied by the differential binding of key transcription factors, resulting in increases or decreases in gene expression.

As shown in Fig. 2B, the surrounding regions of differentially expressed genes were enriched for differential ChIP-seq and ATAC-seq peaks that changed in the same direction as the genes in decidualized samples. Accordingly, when we paired differential peaks with the nearest expressed gene as its putative gene target, we observed that these pairs were more likely to have matching directions of change (i.e., both the peak and the gene have increased or decreased read counts in decidualized samples) than nonmatching directions when compared with pairs that were assigned randomly (Fig. 3A).

(A) Randomly assigning a gene to a peak (see Materials and Methods) resulted in fewer peaks that matched the direction of change with that of differentially expressed genes than when using pcHi-C interactions or the nearest gene to pair peaks to genes. (B) The FOXO1 gene is more highly expressed in decidualized samples (fourfold increase, P = 7 1022) and its promoter physically interacts (red arcs) with distal regulatory elements (yellow highlights) that show increased activation in decidualized samples. The nearest expressed gene to these differential peaks is COG6.

In many cases, however, the target gene for a regulatory element is not the nearest gene (36), and therefore, information about distal chromatin interactions can be useful in prioritizing candidate gene targets of variants identified in GWAS. To this end, we generated a pcHi-C map of a decidualized cell line, thus enriching for the identification of long-range chromatin interactions between promoters and distant regulatory elements (3739). We identified a total of 161,337 interactions, of which 53,211 were between promoters and distal regions of accessible chromatin assayed by ATAC-seq and ChIP-seq, suggestive of their regulatory role. We used the significant interactions between promoters and distal regions that we identified to pair differential peaks with putative target genes. As shown in Fig. 3A, using pcHi-C interactions as a pairing method resulted in enhanced identification of differential peak/differential target gene pairs that have matching directions of change compared to random assignment of gene-target pairs.

Whereas assigning peaks to the nearest expressed gene also led to enhanced assignment of differential peaks to target genes with matching directions of change (Fig. 3A), pcHi-C was helpful in identifying less obvious target genes, as shown in Fig. 3B. In this example, several pcHi-C interactions link distal regulatory elements up to 847 kb away that became more active in decidualized cells to the promoter of a gene (FOXO1) that was up-regulated in decidualized cells and is known to be involved in decidualization (32). The nearest expressed gene method assigned those differential peaks to COG6, a gene that does not change expression in decidualized samples and is therefore a less likely target.

In conclusion, by combining pcHi-C interactions with the epigenome maps and transcriptome data, we were able to identify genes and putative regulatory elements that respond to, or regulate, the decidualization process. We next used these functional genomic maps and datasets to fine map GWAS loci for gestational duration and identify new candidate genes with a potential role in PTB.

To identify candidate genes that may play a role in gestational duration and PTB, we used summary data from a GWAS of gestational duration based on a meta-analysis of a 23andMe GWAS (n = 42,121) (5) and the results from six European datasets (n = 14,263). A detailed description of the GWAS is in the Supplementary Materials and figs. S1 and S2. After filtering for single-nucleotide polymorphism (SNPs) that are present in the 1000 Genomes Project data and minor allele frequency of >0.01, we identified SNPs at six autosomal loci, defined as approximately independent blocks by LDetect (40), that were associated with gestational duration at genome-wide significance of P < 5 108 (table S2). We then created a computational pipeline to assess enrichment of GWAS signals in functional annotations that we generated in untreated (MSCs) and decidualized (DSCs) stromal cells to fine map GWAS loci and discover candidate causal genes and to potentially provide support for additional loci that did not reach genome-wide significance in the GWAS (Fig. 4A). Each step of this procedure is explained below and described in details in Materials and Methods.

(A) Computational pipeline for analyzing GWAS of gestation duration. Yellow boxes (input data): GWAS summary statistics and functional annotations from endometrial stromal cells (in both untreated and decidualized cells). Green boxes: Stages of statistical analysis (see Materials and Methods). (B) Stratified LDSC heritability analysis of GWAS of gestational duration using functional annotations. Left: Fold enrichment of heritability in each annotation. Dashed line shows values at 1, i.e., no enrichment. Center: Proportion of heritability explained by each annotation. Right: Proportion of SNPs across the genome that fall within an annotation. For each annotation, enrichment (left) is the ratio of h2 proportion (center) divided by the SNP proportion (right). Error bars represent 95% confidence intervals.

We first used stratified linkage disequilibrium (LD) score regression (S-LDSC) (41) to assess enrichment of GWAS signals in functional annotations in endometrial stromal cells. S-LDSC takes as input GWAS summary statistics across the genome and functional annotations of SNPs, e.g., whether an SNP is in ATAC-seq peak, and returns as output heritability enrichment of each annotation. S-LDSC is a commonly used tool for estimating the proportion of heritability of complex phenotypes that is explained by variants in certain functional annotations. The heritability enrichment is defined by the proportion of heritability explained by annotations divided by the expected proportion, which is the percent of SNPs genome wide that are in these functional annotations. To account for possible systematic bias in this analysis, i.e., SNPs within annotations of interest may differ from background SNPs in systematic ways such as their LD structure and epigenomic properties, we included a range of baseline annotations (default S-LDSC setting), including LD-related annotations, deoxyribonuclease (DNase) hypersensitivity, enhancer annotation, H3K27ac, H3K4me1, and other histone marks (the union across cell types). Thus, if an annotation is shared by many cell types, then it would not show the enrichment in S-LDSC analysis (see Materials and Methods).

Using S-LDSC, we found 5- to 10-fold enrichments of GWAS heritability for gestational duration in our functional annotations compared to the baseline model of S-LDSC (Fig. 4). The enrichment of enhancer marks H3K27ac and H3K4me1 was higher in decidualized than in untreated cells, but the opposite pattern was observed for the promoter mark H3K4me3, which was more enriched in untreated (MSCs) than in decidualized (DSCs) cells. These findings are consistent with previous observations that enhancers are often more dynamic and condition- or tissue-specific than promoters (10). We observed weaker heritability enrichments of open chromatin regions defined by ATAC-seq and of interaction regions in pcHi-C. However, because we performed joint analysis of all annotations together, the enrichment of one annotation (e.g., ATAC-seq peaks) will be reduced if the enrichment is partially explained by other, overlapping annotations (e.g., H3K27ac). Although the promoter mark H3K4me3 in untreated cells showed the highest enrichment, the annotations that contributed most to the heritability of gestational duration were enhancers (Fig. 4) due to the much larger number of enhancer histone marks than promoters in the genome. Our results thus highlight the importance of functional annotations in endometrial stromal cells at GWAS loci for gestational duration.

We next developed a computational procedure, based on fine mapping, to integrate the decidua stromal cell functional maps with a GWAS of gestational duration to identify putative causal variants (Fig. 4A). Because of extensive LD in the human genome, the causal variants driving the associations are unknown at most loci discovered by GWAS. Fine mapping is a Bayesian statistical procedure that takes as input GWAS summary statistics and patterns of LD at trait-associated loci and computes the probability of each variant at a locus to be a causal variant (7). These probabilities, known as posterior inclusion probabilities (PIPs), reflect our confidence of certain SNPs being causal variants. The PIP of a variant ranges from 0 to 1, with 1 indicating full confidence that the SNP is a causal variant. If a region contains a single causal variant, the PIPs of all SNPs in the region should approximately sum to 1.

While fine mapping has been commonly used in identifying putative causal variants from GWAS of complex traits (7), it is often difficult to narrow down causal signals to one or a small number of variants in most GWAS loci. Standard fine mapping treats all SNPs at a locus equally. Recent work suggests that incorporating Bayesian prior probabilities that favor functional SNPs improves fine mapping (8, 42). We posited that integrating functional annotations in pregnancy-relevant cells in a statistical fine-mapping framework would aid in (i) identifying candidate causal variants at each locus associated with gestation duration, (ii) linking those variants to their target genes, and (iii) discovering additional loci and genes associated with gestational duration that may have failed to reach the stringent threshold for significance in GWAS.

We first leveraged the enrichments of DSC annotations to create Bayesian prior probabilities for a variant being causal. On the basis of the results of S-LDSC, we chose H3K27ac, H3K4me1, and pcHi-C interactions from the decidualized cells, and H3K4me3 from untreated cells, as functional genomic annotations to create informative priors using TORUS (42). TORUS takes as input genome-wide summary statistics from GWAS and the functional annotations of SNPs and computes enrichment parameters of annotations, which reflect how much more likely an SNP is a causal variant than randomly chosen SNPs (table S3). SNPs associated with functional annotations are generally assigned higher prior probabilities. In addition, TORUS computes statistical evidence at the level of genomic blocks, defined as the probability that a block (determined by LD) contains at least one causal SNP. Without including any histone marks or chromatin accessibility annotations, TORUS implicated six autosomal blocks in the genome at false discovery rate (FDR) of < 0.05, including five of the six genome-wide significant autosomal loci identified in the GWAS (P < 5 108). One locus on chromosome 3 had an FDR = 0.11 and was therefore not identified by TORUS, and one locus on chromosome 9 that was not identified in the GWAS was implicated by TORUS (data file S3). By including the functional genomic annotations from endometrial stromal cells, the number of high confidence blocks increased to 10, including all 6 that were significant in the gestational duration GWAS and 4 that were not significant in the GWAS (data file S3).

We next performed computational fine mapping on these 10 blocks, with the informative priors learned by TORUS, using sum of single effects (SuSiE) regression (43). Conceptually, SuSiE is a Bayesian version of the stepwise regression analysis commonly used in GWAS (i.e., conditioning on one variant and testing if there is any remaining signal in a region). SuSiE accounts for the uncertainty of causal variants in each step and reports the results in the form of PIPs. Including the priors defined by TORUS using DSC functional annotations significantly improved fine mapping (Fig. 5A, table S3, and data file S4). For example, only one SNP reached PIP > 0.3 across all 10 blocks using the default setting under SuSiE (uniform prior, treating all SNPs in a block equally). This reflects the general uncertainty of pinpointing causal variants due to LD, e.g., a strong GWAS SNP in close LD with nine other SNPs would have PIP about 0.1. By using the annotation-informed priors, eight SNPs in six different blocks reached PIP > 0.3 (Fig. 5A). In some blocks, we were able to fine-map a single high-confidence SNP, e.g., the FOXL2 locus on chromosome 3, while in other blocks, we had considerable uncertainty of the causal variants, as shown by large credible sets, i.e., the minimum set of SNPs to include the causal SNP with 95% probability (Fig. 5B). Table 1 summarizes the most probable causal variants in eight blocks (fine mapping in the remaining two blocks produced large credible sets with no high-PIP SNPs) and their likely target genes based on promoter assignment or chromatin interactions from pcHi-C. We note that our results of the WNT4 locus identified rs3820282 as the likely causal variant. This is consistent with our previous results demonstrating experimentally that the T allele of this SNP disrupts the binding of estrogen receptor 1 (5). This SNP was among the three most likely SNPs in our fine-mapping study, with a PIP of 0.27 (Table 1).

(A) PIPs of SNPs using uniform vs. functional priors in SuSiE (each dot is an SNP). The functional prior of an SNP is based on SNP annotations and is estimated using TORUS. (B) Summary of fine-mapping statistics of all 10 regions. X axis: The size (number of SNPs) of credible set. Y axis: The maximum PIP in a region. We label each region by its top SNP (by PIP) and the likely causal gene, according to Table 1 or the nearest gene of the top SNP. (C) Likely causal variants near HAND2 and their functional annotations. The top panel shows the significance of SNP association in the GWAS and the middle panel shows fine-mapping results (PIPs) in the region. The vertical yellow bar highlights the two SNPs with high PIPs. These SNPs are located in a region annotated with ATAC-seq, H3K27ac, H3K4me1, and H3K4me3 peaks (bottom). This putative enhancer also had increased ATAC-seq, H3K27ac, and H3K4me1 levels in decidualized samples and interacts with the HAND2 promoter (red arc).

Functional annotations are based on data from endometrial stromal cells. We list an annotation if the SNP is located in a sequence with that annotation in either untreated or decidualized condition. Functional prior is the prior probability of an SNP being a causal variant. For an SNP without any functional annotation, its prior probability is 3.6 106. We list the pcHi-C annotation if the SNP is within 1 kb of a region involved in a pcHi-C interaction. We call a gene the target of an SNP if (i) the SNP is located in the promoter (< 1 kb of transcription start site) of that gene or (ii) the promoter of that gene has a pcHi-C interaction with a region within 1 kb of the SNP. In the case of rs147843771 at the FOXL2 locus, the target was defined by literature evidence (69). The number of credible SNPs at each region is shown in Fig. 5B. SNPs in bold are discussed in the text. FOXL2 (69), forkhead box L2; GATA2, GATA-binding protein 2; HAND2, heart and neural crest derivatives expressed 2; KCNAB1, potassium voltage-gated channel subfamily A member regulatory beta subunit 1; WNT4, Wnt family member 4.

We highlight the results from two regions. In both cases, we were able to identify putative risk genes with relatively high confidence, and neither is the nearest gene of lead SNPs in GWAS. In the first case, two adjacent SNPs [311base pair (bp) apart], rs13141656 and rs7663453, on chromosome 4q34 did not reach genome-wide significance in the GWAS (P = 3.9 107 and 4.5 107, respectively). After using functional annotations in decidua-derived stromal cells, the block containing these SNPs was highly significant (TORUS q = 0.02), suggesting the presence of at least one causal variant in this block. The two SNPs together explained most of the PIP signal in the block (PIP 0.38 and 0.33, respectively, Table 1). The two SNPs are located in a region of open chromatin in endometrial stromal cells, with enhancer activity marked by both H3K27ac and H3K4me1 (Fig. 5C). Only 9 of the 129 tissues from the Epigenome Roadmap (11) also had H3K27ac, H3K4me1, or H3K4me3 peaks spanning the rs13141656 locus and only 2 spanning the rs7663453 locus. In addition, this putative enhancer is bound by multiple transcription factors, including GATA2, FOXO1, NR2F2, and PGR, based on ChIP-seq data. The only physical interaction of this enhancer in the pcHi-C data in decidualized stromal cells is with the promoter of the HAND2 gene, located 277 kb away (Fig. 5C). Summing over the PIPs of all SNPs whose nearby sequences interact with HAND2 (heart and neural crest derivatives expressed 2) via chromatin looping gives an even higher probability, 0.89, suggesting that HAND2 is very likely to be the causal gene in this region (table S4). HAND2 is an important transcription factor that mediates the effect of progesterone on uterine epithelium (44). Thus, in this example, we identified a previously unknown locus, the likely causal variant(s), the enhancers they act on, and an outstanding candidate gene for gestational duration and PTB.

The second example focuses on the locus showing a strong GWAS association with gestational duration on chromosome 3q21. The lead SNP, rs144609957 (GWAS P = 4 1013), is located upstream of the EEFSEC (eukaryotic elongation factor, selenocysteine-tRNAspecific) gene. There is considerable uncertainty of the causal variants in this region, with 50 SNPs in the credible set and the lead SNP explaining only a small fraction of signal (PIP = 0.02). Among all 12 SNPs with PIP > 0.01, 11 have functional annotations, most commonly H3K4me1 and pcHi-C interactions. For nine SNPs (first three shown in Table 1), the sequences in which they are located physically interact with the promoter of GATA2 in the pcHi-C data but not with any other promoters in the region (fig. S3). The PIPs of all SNPs in the genomic regions that likely target GATA2 through chromatin looping sum to 0.68 (table S5). Thus, despite uncertainty of causal variants in this region, our results implicate GATA2 as a candidate causal gene in endometrial stromal cells. GATA2 is a master regulator of embryonic development and differentiation of tissue-forming stem cells (45). As support for the possible role of GATA2 in pregnancy, GATA2 deficient mice show defects in embryo implantation and endometrial decidualization (35), making this another excellent candidate causal gene for gestational duration and PTB.

The molecular processes that signal the onset of parturition in human pregnancies, and how perturbation of those processes result in PTB, are largely unknown. Yet, understanding these processes would reveal important insights into the potential causes of adverse pregnancy outcomes, including spontaneous labor before 37 weeks gestation, and potentially lead to the identification of biomarkers and therapeutic targets for PTB. Although it is experimentally challenging to link decidualization processes directly to parturition in humans, it is well accepted that shallow implantation due to suboptimal decidualization is associated with poor pregnancy outcomes in general (4648) and that the decidua is key in triggering parturition (13, 14). Thus far, however, specific genes that perturb decidualization processes and lead to PTB are poorly defined.

Unbiased GWASs do not require prior knowledge of molecular processes underlying disease phenotypes and have the potential to identify novel genes and pathways contributing to common diseases. However, the significant heterogeneity of most common diseases and small effects of most common disease-associated variants lead to the requirement for very large sample sizes (in the tens to hundreds of thousands of cases) to discover more than a handful of associated loci that meet stringent criteria for genome-wide significance. To address this limitation and provide orthogonal evidence for assessment of associations, we characterized the transcriptional and chromatin landscapes in decidua-derived stromal cells and integrated those functional annotations with a GWAS of gestational duration to discover novel loci and genes. The primary motivation for these studies was the notable paucity of genomic and epigenomic functional annotations in pregnancy-relevant primary cells among those studied by large consortia (911). Here, we filled a significant gap by providing maps in untreated and decidualized stromal cells and used these maps for annotating GWAS of pregnancy-related traits.

We chose to focus these studies on endometrial stromal cells because of their central importance in both the establishment and maintenance of pregnancy, as well as their intimate juxtaposition to fetal trophoblast cells throughout pregnancy. Of particular relevance are the roles that decidualized stromal cells play in regulating trophoblast invasion, modulating maternal immune and inflammatory responses at the maternal-fetal interface, and controlling remodeling of the endometrium (48). Defects in all of these processes have been considered a contributing factor to pregnancy disorders (48, 49). Moreover, we showed that the SNPs in regions with endometrial stromal cell functional annotations explained more of the heritability of gestational duration compared to just using baseline annotations. Among all annotations, enhancer marks H3K4me1 (in both decidualized and untreated stromal cells) and H3K27ac (in decidualized cells) were 8- to 10-folds enriched at GWAS loci after adjusting for the general annotations and accounted for 50 to 70% of the GWAS heritability. The lack of complete independence between these marks makes it difficult to delineate their individual effects but, nonetheless, highlights the importance of enhancers and of gene regulation in endometrial stromal cells in modulating the effects of GWAS variants on gestational duration. This is consistent with both the known tissue-specific roles of enhancers and the observation that more than 90% of GWAS loci reside outside of the coding portion of the genome and are enriched in regions of open chromatin and enhancers (12, 41).

Integrating transcriptional and chromatin annotations of gene regulation from MSCs and DSCs improved our ability to discover novel GWAS loci and identify likely causal SNPs and genes associated with gestational duration. We illustrate how our integrated platform identified a novel causal locus and candidate gene (HAND2) associated with gestational duration, as well as refined the annotation of loci that had been previously identified. Our data suggest that in endometrial stromal cells, GATA2 is likely the target gene of enhancers harboring SNPs associated with gestational duration. This does not exclude the possibility that the nearest gene to the associated SNPs, EEFSEC, may be a target gene in other cell types. Both HAND2 (50) and GATA2 (51) are involved in decidualization processes in humans, and perturbations in this process have been linked to poor pregnancy outcomes (4648). Neither GATA2 nor HAND2 was identified as potential candidate genes in previous GWASs of gestational duration, or PTB supports our approach and the importance of using functional annotations from cell types relevant to pregnancy to fine map and identify candidate genes for the pregnancy-related traits. Overall, the integrated analyses performed in this study resulted in the identification of both novel GWAS loci and novel candidate genes for gestational duration, as well as maps of the regulatory architecture of these cells and their response to decidualization.

However, there are some limitations. Our results are based on cells from only three individuals, which may not fully capture the regulatory landscape of endometrial stromal cells. For pcHI-C, we used cells from a single individual to generate the chromatin interactions map. Another limitation is that we focused on only one cell type, albeit one that plays a central role in pregnancy and only one exposure (hormonal induction of decidualization) at one time point (48 hours). Furthermore, it is unclear how our model of in vitro decidualization mimics the endogenous decidualization of endometrial cells during pregnancy. While we chose decidualization as a perturbation to ascertain the dynamic features of functional genomic annotations, we fully anticipate that obtaining annotations in other cell types and in response to other relevant perturbations will improve the ability to identify novel loci, variants, and genes associated with PTB. Future studies that include fetal cells from the placenta and uterine or cervical myometrial cells could reveal additional processes that contribute to gestational duration and PTB, such as those related to fetal signaling and the regulation of labor, respectively. Inclusion of additional exposures, such as trophoblast conditioned media (52) and additional exposure times, may further reveal processes that are pregnancy specific. Second, to maximize power, we focused on a GWAS of gestational duration and not PTB per se. While previous GWAS have shown that all PTB loci were among the gestational age loci (5), we realize that some of the loci that we identified could be related to normal variation in gestational duration and not specifically to PTB. Nonetheless, our findings contribute to our understanding of potential mechanisms underlying the timing of human gestation, about which we still know little. Last, although our ChIP-seq results revealed an association between GATA2 binding and decidualization, confirming the role of this transcription factor in decidual cell biology (53, 54), and studies in murines support its role in endometrial processes (35), we do not yet have direct evidence showing that perturbations in the expression of GATA2, or any of the other target genes identified, influence the timing of parturition in humans. Future studies will be needed to directly implicate the expression of these genes in gestational duration or PTB. Our study highlights the importance of generating functional annotations in pregnancy-relevant cell types to inform GWASs of pregnancy-associated conditions. Our results suggest that the expression of two transcription factors, GATA2 and HAND2, in endometrial stromal cells may regulate transcriptional programs that influence the timing of parturition in humans, which could lead to the identification of biomarkers of or therapeutic targets for PTB.

This study was approved by the Institutional Review Boards at the University of Chicago, Northwestern University, and Duke University Medical School. Informed consent for the use of data collected via questionnaires and clinics was obtained from participants following the recommendations of the ALSPAC (Avon Longitudinal Study of Parents and Children) Ethics and Law Committee at the time. Informed consent for the use of genetic data in the other six GWASs used in this study was also obtained from participants. Details are available in the Supplementary Materials.

Placentas were collected from three African American women (18 years old) who delivered at term (37 weeks) following spontaneous labor; all were vaginal deliveries of singleton pregnancies. Within 1 hour of delivery, 5 cm by 5 cm pieces of the membranes were sampled from a distant location of the rupture site. Pieces were placed in Dulbeccos modified Eagles medium (DMEM)-Hams F12 media containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin. Samples were kept at 4C and processed within 24 hours of tissue collection. This study was approved by the Institutional Review Boards at the University of Chicago, Northwestern University, and Duke University Medical School.

Third trimester placental tissue was enzymatically digested by a modification of previously described methods (55, 56). Decidua tissue was gently scraped from chorion, and tissue was enzymatically digested in a solution (1 Hanks balanced salt solution, 20 mM Hepes, 30 mM sodium bicarbonate, and 1% bovine serum albumin fraction V) containing collagenase type IV (200 U/ml; Sigma-Aldrich, C-5138), hyaluronidase type IS (1 mg/ml; Sigma-Aldrich, H-3506), and DNase type IV (0.45 KU/ml, Sigma-Aldrich, D-5025) at 37C, until a single-cell suspension was obtained (usually three rounds of 30 min digestion using fresh digestion media each round). Epithelial cells were removed by filtering through a 75 M nylon membrane and RPMI (Sigma-Aldrich) containing 10% FBS was added for enzyme inactivation. Dissociated cells were collected by centrifugation at 400g for 10 min and washed in RPMI/10% FBS. Erythrocytes were removed by cell pellet incubation with 1 red blood cell lysis buffer (Sigma-Aldrich) for 2.5 min at room temperature. The resulting cells were counted and resuspended in seeding media [1 phenol red-free high-glucose DMEM (Gibco)] supplemented with 10% FBS (Thermo Fisher Scientific), 2 mM l-glutamine (Life Technologies), 1 mM sodium pyruvate (Fisher Scientific), 1 insulin-transferrin-selenium (ITS; Thermo Fisher Scientific), 1% penicillin/streptomycin, and 1 antibiotic-antimycotic (Thermo Fisher Scientific). Dissociated cells were plated into a T75 flask and incubated at 37C and 5% CO2 for 15 to 30 min (enrichment by attachment). The supernatant was carefully removed, and loosely attached cells were discarded. Plates were allowed to grow in fresh media containing 10% charcoal-stripped FBS (CS-FBS), and 1 antibiotic-antimycotic until the plate was 80% confluent. The antibiotic-antimycotic was removed from the culture media after 2 weeks of culture. We obtained >99% vimentin-positive cells after three passages (fig. S4). Cells were expanded, harvested in 0.05% trypsin, and cryopreserved in 10% dimethylsulfoxide culture media for subsequent use. Each cell line was defined as coming from a different sample collection (different pregnancy).

Cells were plated and grown for 2 days in cell culture media (1 phenol red-free high-glucose DMEM, 10% CS-FBS, 2 mM l-glutamine, 1 mM sodium pyruvate, and 1 ITS). After 2 days, cells were treated either with control media (1 phenol red-free high-glucose DMEM, 2% CS-FBS, 2 mM l-glutamine) or decidualization media (1 phenol red-free high-glucose DMEM, 2% CS-FBS, 2 mM % l-glutamine, 0.5 mM 8-Br-cAMP, and 1 M MPA) for 48 hours. Cells were incubated at 37C and 5% CO2 and harvested for ATAC-seq, ChIP-seq, and RNA-seq, and prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP1) mRNA were assessed by quantitative real-time polymerase chain reaction (PCR) before each downstream assay was performed.

Total RNA was extracted from approximately 1 million cells using the AllPrep DNA/RNA Kit (QIAGEN) according to manufacturers instructions. RNA quality (RNA integrity number) and concentration was assessed by Bioanalyzer 2100 (Agilent technology). RNA-seq libraries were generated by a TruSeq stranded total RNA library prep kit (Illumina) and TruSeq RNA CD Index Plate.

For ChIP experiments, cells were cross-linked by adding to the media 37% formaldehyde to a final concentration of 1%, gently mixed, incubated for 10 min, and quenched for 5 min with 2.5 M glycine for a final of 0.125 M per plate. Cells were washed using cold 1 phosphate-buffered saline and scraped in 15 ml of cold Farnham lysis buffer and protease inhibitor (Roche, 11836145001), and cell pellets were flash frozen and kept at 80C. Thawed pellets were resuspended in radioimmunoprecipitation assay buffer on ice, aliquoted into 20 million cells per tube, and sonicated by Bioruptor (three 15-min rounds of 30 s ON, 30 s). ChIP was performed on 10 million cells using antibodies to H3K27ac, H3K4me3, and H3K4me1 histone marks (ab4729/lot no. GR274237, ab8580/lot no. GR273043, and ab8895/lot no. GR262515, respectively). M-280 sheep anti-rabbit immunoglobulin G Dynabeads (Invitrogen, 11203D) was used for chromatin immunoprecipitation. DNA was purified using the Qiagen MinElute PCR Purification Kit, quantified by Qubit, and prepared for sequencing using the Kapa Hyper Prep Kit. All libraries were pooled to 10 nM per sample before sequencing.

Approximately, 50,000 cells were harvested and used for ATAC-seq library preparation as described in the Fast-ATAC protocol (57). ATAC-seq libraries were uniquely indexed with Nextera PCR Primers and amplified with 9 to 12 cycles of PCR amplification. Amplified DNA fragments were purified with 0.8:1 ratio of Agencourt AMPure XP (Beckman Coulter) to sample. Libraries were quantified by Qubit, and size distribution was inspected by Bioanalyzer (Agilent Genomic DNA chip, Agilent Technologies). All libraries were pooled to 10 nM per sample before sequencing.

In situ Hi-C was performed as described previously (58). Briefly, 5 million decidualized cells were treated with formaldehyde 1% to cross-link interacting DNA loci. Cross-linked chromatin was treated with lysed and digested with MboI endonuclease (New England Biolabs). Subsequently, the restriction fragment overhangs were filled in and the DNA ends were marked with biotin-14-dATP (Life Technologies). The biotin-labeled DNA was sheared and pulled down using Dynabeads MyOne Stretavidin T1 beads (Life Technologies, 65602) and prepared for Illumina paired-end sequencing. The in situ Hi-C library was amplified directly off of the T1 beads with nine cycles of PCR using Illumina primers and protocol (Illumina, 2007). Promoter capture was performed as described previously (39). The Hi-C library was hybridized to 81,735 biotinylated 120-bp custom RNA oligomers (Custom Array) targeting promoter regions (four probes/RefSeq transcription start sites). After hybridization, postcapture PCR was performed on the DNA bound to the beads via biotinylated RNA.

Read counts per gene were calculated with Salmon (59) version 0.12.0 on transcripts from human Gencode release 19 (ftp://ftp.ebi.ac.uk/pub/databases/gencode/Gencode_human/release_19/gencode.v19.pc_transcripts.fa.gz and ftp://ftp.ebi.ac.uk/pub/databases/gencode/Gencode_human/release_19/gencode.v19.lncRNA_transcripts.fa.gz). Estimated counts were used in exploratory analysis (transformed with DESeq2s rlog function) and in DESeq2 (25) version 1.24.0 to identify differentially expressed genes (adjusted P 0.05 and absolute fold change of 1.2). After observing that replicates for each cell lines clustered together, we pooled reads for each cell line, combining three decidualization experiments in each sample. We then performed a paired analysis to obtain genes that were differentially expressed between untreated and decidualized samples. The six samples clustered by treatment and by cell line and analysis with svaseq (60) showed that the two surrogate variables identified correlated with cell line, and therefore, a paired analysis was enough to correct the data.

Similarly to RNA-seq, we pooled reads from replicates for each cell line. We called peaks for each of the six samples using MACS2 and converted peak coordinates into 100-bp contiguous bins. Bins covered by less than 60% of their extension were excluded. To identify reproducible peaks, we only kept bins that were present in at least two of the three cell lines in each condition, allowing for condition-specific peaks. See table S7 for an assessment of the contribution of each cell line to the universe of peaks obtained. We then merged all adjacent bins, expanding them back into longer peaks. We counted the number of reads in all peaks and in all samples and compared the read counts using DESeq2 (adjusted P < 0.05 and absolute fold change >1.2).

The P values in Fig. 2B were calculated with a chi-square test of the number of peaks with increased or decreased numbers of reads observed and an expected probability based on the number of peaks in each category for each dataset. Bonferroni correction was performed to correct for multiple testing.

ChIP-seq reads were downloaded from National Center for Biotechnology Information Gene Expression Omnibus and processed locally. HOMER 4.9 (63) was used to call peaks for the following samples: PGR (GSE94038); NR2F2 (GSE52008); FOSL2 (GSE94038), FOXO1 (GSE94037); and NR2F2 input (GSE52008); and FOXO1, PGR, and FOSL2 input (GSE94038).

Reproducible peaks were converted into 100 bp bins and those with >60% of their extension covered by a peak was retained. Common bins were counted, and the number of counts was plotted with UpSetR 1.4.0.

We used HOMER 4.9 to identify DNA binding motifs enriched in peaks with parameters -len 8,10,12 -size 200 -mask.

Enrichment was calculated as the observed number of overlapping peaks divided by the expected number of overlapping peaks using bedtools intersectBed with a 1 bp minimum. The expected number of overlapping peaks was obtained by averaging 100 random samples of peaks with bedtools shuffle excluding gaps annotated by the University of California, Santa Cruz Genome Browser (64). While shuffling peaks does not account for mapping and other biases that make peak locations nonuniform and may result in overestimation of enrichment, our results are limited to comparisons between enrichments, which should cancel any biases.

We used HiCUP v0.5.9 (65) to align and filter Hi-C reads. HiCUP used bowtie2 version 2.2.3 to align reads. Unique reads were used as input by CHiCAGO (66) version 1.2.0, and significant interactions were called with default parameters. We only kept interactions identified by CHiCAGO that were in cis and with an end located at least 10 kb from a capture probe.

To pair peaks using pcHi-C, significant interactions identified by CHiCAGO that overlapped an ATAC-seq or ChIP-seq peak and were less than 300 kb away from a promoter were used. We chose 300 kb because the mean distance between interacting promoters and other regions was 280 kb (median, 200 kb). To pair peaks to the nearest gene, BEDTools closest -t first -d was used to find the gene closest to a peak, up to 300 kb away. To pair peaks to a random gene, all genes up to 300 kb from a peak were selected and one gene was randomly assigned to each peak. For each of these sets of pairs, we calculated the fraction of peak/gene pairs that had the same direction of change according to differential read count analysis with DESeq2, of the total number of peak/gene pairs. Only genes expressed at >1 transcript per million across all samples were used in the nearest and random gene assignments.

P values were calculated with a chi-square test comparing the number of cases in the matched and unmatched categories observed in the random set (average from 200 iterations) and in the two peak/gene pairing methods: nearest gene and pcHi-C interactions.

The GWAS results used in this study was an extension of our previously published results (5). Like our previous study, we used summary results from 23andMe, which were obtained from GWAS of gestational duration in 42,121 mothers of European ancestry. In addition, we performed GWA analyses in 14,263 European mothers from six academic datasets. To increase the power of GWA discovery, we performed meta-analysis between the results from 23andMe and the results from the six datasets. See the Supplementary Materials for a full description of the GWAS.

We assessed how much of the heritability of gestational duration is contained within ATAC-seq, H3K4me1, H3K4me3, H3K27ac, and pcHi-C peaks using S-LDSC (41). S-LDSC is a generalization of LD score regression, a method for estimating the heritability of a trait using SNP-level GWAS summary statistics and SNP-level estimates of the amount of genetic variation tagged at each variant, known as LD scores. Under the LD score regression model, the expected value of the GWAS summary statistic for a variant (specifically, the expected value of the 2 statistic) is a linear function of the LD score at that site, and h2, the per-SNP heritability, and a an intercept parameter. Under the S-LDSC model, rather than estimating a single per-SNP heritability parameter, a parameter is estimated for each of several functional annotations. In a standard S-LDSC analysis, user-provided annotations are combined with a baseline set of genomic annotations from publicly available datasets. For this analysis, LD scores were calculated using the peaks identified as reproducible across either treated or untreated samples as annotations and the genotype data from the European individuals from phase 3 of the 1000 Genomes project (obtained from the Price Lab website: https://alkesgroup.broadinstitute.org/LDSCORE/) as a reference LD panel, using only the HapMap3 SNP list (also from the Price lab website). S-LDSC was performed on the gestational duration GWAS using the endometrial-tissue derived LD scores and the baseline LD scores contained in version 2.2 of the LD score regression baseline LD model. We include all annotations from the baseline LD model except those flanking annotations. This resulted in a total of 64 baseline annotations used in our S-LDSC analysis.

Fine mapping proceeded in three stages. In the first stage, we partitioned the genome into 1703 regions approximately independent regions using breakpoints derived by Berisa et al. (40). Next, we constructed an SNP-level prior probability of being causal variant, informed by the functional genomic data that we collected. We used a Bayesian hierarchical model [TORUS (42)]. TORUS takes as input GWAS summary statistics and genomic annotations and estimates the extent to which SNPs with functional genomic annotations are likely to be causal for a trait of interest. Specifically, under TORUS, each SNP has a small prior probability of being a causal variant, which is a logistic function of the annotations of the SNP. Then, TORUS estimates the parameters of this logistic function using genome-wide summary statistics. Once these parameters are estimated, each SNP will have a prior causal probability based on its unique functional annotations. We ran TORUS with the gestational age GWAS summary statistics and the reproducible H3K27ac and H3K4me1 peaks from the treated samples along with the pcHi-C contact regions to obtain an SNP-level prior.

Last, fine mapping was performed using a summary statistics-based version of the sum of single effects model (43) using 1000 Genome as reference panel. SuSiE (as implemented in the R package susieR) was run on the 10 regions believed to have one or more causal variants with an FDR of 0.1 as estimated by TORUS. For each region, SuSiE was run with a uniform prior (default setting of SuSiE) and with an informed prior learned by TORUS. The parameter L of SuSiE (maximum number of causal variants) is set at 1 when running SuSiE (67, 68).

H3K27ac, H3K4me1, and H4K4me3 histone modification peak coordinates were downloaded from the Epigenome Roadmap data website, and bedtools intersect was used to find peaks that overlapped SNPs coordinates.

Read the rest here:
Transcriptome and regulatory maps of decidua-derived stromal cells inform gene discovery in preterm birth - Science Advances

Read More...

Therapy Options Compared for the Treatment of Recurrent DLBCL – Targeted Oncology

December 4th, 2020 12:16 am

Loretta J. Nastoupil, MD, associate professor, director, Lymphoma, Outcomes Database Section chief, New Drug Development Department of Lymphoma/Myeloma ,Division of Cancer Medicine at The University of Texas MD Anderson Cancer Center, discussed the use of chimeric antigen receptor (CAR) T-cell therapy in patients with diffuse large B-cell lymphoma.

The discussion occurred among a group of oncologist during a Targeted Oncology Case Base Peer Perspective event.

Targeted Oncology: What clinical trial data support the use of CAR T-cell therapy for this patient?

NASTOUPIL: The ZUMA-1 study [NCT02348216] enrolled patients with refractory large cell lymphoma and included about 30% of patients with double-hit features. [It] also included [patients with] primary mediastinal transformed follicular lymphoma. There was a relatively small sample size of 101 patients. This was a single-arm, phase 2 [design]. However, they had a high success rate in terms of enrollment and treatment of patients where 91% of the patients who were enrolled received cell therapy.

ZUMA-1 [examined axicabtagene ciloleucel (axi-cel; Yescarta)], an autologous CD19-directed CAR T-cell therapy. It has a CD28 costimulatory molecule. The lymphocyte depleting regimen is high in this study: 500 mg/m2 of cyclophosphamide and 30 mg/m2 of fludarabine for 3 consecutive days.

The median progress-free survival [PFS] in this study was about 6 months [5.9 months; 95% CI, 3.3-15.0]. The best objective response rate [ORR] was 83% and best complete response [CR] rate was 58%.

There was a stark drop-off and then a flattening of the [Kaplan-Meier PFS] curve. [Experience has shown that] theres about 40%, to maybe as high as 50%, of patients who can be cured with this, but its hard to identify who they are. The patients who dont respond generally do poorly within the first 3 to 6 months.

The JULIET study [NCT02445248] was a bit different. Its a larger multicenter study [that examined tisagenlecleucel (Kymriah)]. It was a much longer time from enrollment to infusion of CAR T cells; as a result, bridging therapy was allowed. They also included relapsed, not just refractory, patients, and they excluded patients with primary mediastinal disease.2

There [were] lower response rates [in] this study population of 52%. The CR rate, though, is notable at 40%. The patients who had CR had durability of that CR. About 40% of the patients had a meaningful outcome with this CAR-T cell therapy, which was a 4-1BB construct. This is an autologous [product] with a CD19 target.

The efficacy is hard to compare across the studies because they were different in terms of patient eligibility and trial conduct. However, my general sense is that the vast majority of patients have about a 40%, to maybe as high as 50%, meaningful CR that is durable, and the overall survival [OS] is quite notable. With ZUMA-1, the median OS settles in at 27.1 months.

Are there any CAR T-cell therapies that may be added to the list of available products for these patients?

Liso-cel [lisocabtagene maraleucel] is the third [CAR T-cell product that is] anticipated for FDA approval. Thats also a 41BB construct similar to tisagenlecleucel. Its different in that theres a fixed CD4 to CD8 ratio and a longer time for manufacturing in comparison to axi-cel. But again, there is meaningful efficacy.3

How does the toxicity compare between these agents?

The toxicity looks to be different, but different grading systems were applied in the studies. When you apply the same rating system retrospectively, patients have fewer grade 3 or higher cytokine release syndrome in the JULIET study than what was initially reported.4

Axi-cel tends to have the highest rate of grade 3 or higher neurotoxicity and liso-cel tends to have the most favorable toxicity profile. Though because its the third [agent to become available], weve [become] much better at identifying and mitigating some of these acute toxicities.

There are differences, in my opinion, across the 3 constructs in terms of safety, despite the efficacy being quite similar.

Why are these data important to review?

We know CAR-T cell therapy has transformed outcomes for about 40% of patients; however, its logistically challenging....Weve struggled with identifying what makes a good CAR-T cell candidate outside of having chemotherapy-refractory disease and being in a third-line setting.

What other therapy options could be used in this patient if she is not a candidate for CAR T-cell therapy?

Polatuzumab vedotin [Polivy] in combination with bendamustine and rituximab [BR] was approved based on a randomized phase 2 study [NCT02257567].5 Of note, this was a study that included both follicular and large cell lymphoma. However, the follicular [cohort showed] a negative result. There was no significant impact with the addition of polatuzumab vedotin to BR, which is not necessarily surprising given that bendamustine is a very active agent for follicular lymphoma.

It is not an active agent in large cell lymphoma. One of the reasons why it was included in this study design is because the potential for challenge with polatuzumab was identifying an agent in third-line large cell lymphoma setting where you wouldnt have additive.

Please describe the trial that led to the approval of polatuzumab vedotin.

For baseline characteristics, and this is specifically looking at the patients with large cell lymphoma, the median age was 67 years [range, 33-86] in the polatuzumab/BR arm versus 71 years [range, 30-84] in the BR-only arm. In terms of median number of prior lines of therapy, it was 2 for both arms. In terms of prior stem cell transplant, about 25% had undergone transplant in the polatuzumab-BR arm versus 15% in the BR-arm.

ORR by independent review with the polatuzumab-BR was much higher versus the BR-only arm [45% versus 17.5%].6

Importantly, the median PFS was significantly different. If patients received polatuzumab, the median PFS was 9.5 months versus 3.7 months in the control arm [0.36; 95% CI, 0.21-0.63; P < .001].

The important message is that there werent any subgroups that benefited from BR alone, and this included criteria such as bulky disease, number of prior lines of therapy, and duration of response with the prior line of therapy.

The OS was significantly better if you had polatuzumab plus BR, with a median of 12.4 months versus 4.7 months [HR, 0.42; 95% CI, 0.24-0.75; P = .002]. Similarly, there were no subgroups that tended to benefit from receiving BR alone.

What was the toxicity profile of this regimen for patients?

[It is] important to note that peripheral neuropathy, which is an adverse event associated with this antibody-drug conjugant, was mostly reported as grade 1 events. [The effect was] slightly additive if you had the BR and polatuzumab versus no polatuzumab.

In terms of neutropenia, grade 3/4 events with the combination occurred in 46.2% of patients, and thats higher when compared with BR alone [33.3%]. However, febrile neutropenia events were no different between arms [10.3% vs 12.8%].

References:

1. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531-2544. doi:10.1056/NEJMoa1707447

2. Schuster SJ, Bishop MR, Tam CS, et al; JULIET Investigators. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380(1):45-56. doi:10.1056/NEJMoa1804980

3. Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839-852. doi:10.1016/S0140-6736(20)31366-0

4. Abramson JS. Anti-CD19 CAR T-cell therapy for B-cell non-Hodgkin lymphoma. Transfus Med Rev. 2020;34(1):29-33. doi:10.1016/j.tmrv.2019.08.003

5. FDA approves polatuzumab vedotin-piiq for diffuse large B-cell lymphoma. FDA. June 10, 2019. Accessed November 13, 2020. https://bit.ly/2IBZGrv

6. Sehn LH, Herrera AF, Flowers CR, et al. Polatuzumab vedotin in relapsed or refractory diffuse large B-cell lymphoma. J Clin Oncol. 2020;38(2):155-165. doi:10.1200/JCO.19.00172

Link:
Therapy Options Compared for the Treatment of Recurrent DLBCL - Targeted Oncology

Read More...

Retracing the evolutionary emergence of thymopoiesis – Science Advances

December 4th, 2020 12:16 am

INTRODUCTION

The adaptive immune system arguably represents one of the major evolutionary novelties that distinguish vertebrates from their nonvertebrate ancestors. About half a billion years ago, specialized cell types, such as lymphocytes, and new organs, such as the thymus, emerged, contributing to the radical redesign of animal immunity (13). These morphological and cytological innovations likely occurred in the ancestor common to all vertebrates, since they are present in both of the two extant groups of vertebrates, the jawless and jawed vertebrates (3). Additional novelties, such as the chemokine and chemokine receptor systems, enabled new types of cellular interactions, for instance, between hematopoietic progenitor cells and the emerging thymopoietic tissue (4). The thymic microenvironment provides distinct molecular cues such as Notch ligands, chemokines, and cytokines that interact in a synergistic, context-dependent, and hierarchical manner and, in this way, determine the outcome of hematopoietic precursor cell differentiation (5). The key importance of the thymic microenvironment for T cell development was first revealed through studies of rodents homozygous for mutations at the nude locus (68), which disrupt thymus development, causing animals to be immune deficient. The nude locus encodes a transcription factor of the forkhead class (9), now designated as Foxn1. Subsequent studies in mice indicated that the Foxn1 gene is dispensable for the formation of the thymic anlage during embryonic development (8) but is required for the subsequent steps of differentiation of primitive precursor cells into the typical cortical and medullary subsets of the thymic epithelium (10). Foxn1-expressing thymic epithelial precursors (11) are bipotent (12, 13), with each cell able to give rise to a self-organizing thymopoietic unit containing cortical and medullary compartments capable of supporting T cell development (12). Thus, in the absence of Foxn1, the master regulator of the thymic epithelium in mammals, thymus differentiation is aborted, and T cell development fails (9). Attempts to identify the key functional elements of the thymic epithelium through transgenic expression of candidate genes has highlighted the roles of the chemokines Cxcl12 and Ccl25, the cytokine/stem cell factor Scf, and the Notch ligand Dll4 (5), which direct the early steps of T cell development in the thymic microenvironment. However, the individual components of the genetic network downstream of Foxn1 that regulates the emergence of distinct subsets of thymic epithelial cells (TECs) are unknown.

So far, it has not been possible to reconstruct the nature and evolutionary sequence of the steps that gave rise to a functional thymus, as no extant species representing intermediate phylogenetic stages along the transition from innate to adaptive immunity are available for analysis. To begin to address this problem, we have developed an in vivo reconstruction strategy that rests on the exchange of the mammalian version of the Foxn1 transcription factor with its evolutionarily distant relatives of nonvertebrate and vertebrate origin. Since the decisions controlling alternative differentiation pathways in the hematopoietic system are strongly influenced by extrinsic cues (5, 1417), we hypothesized that it should be possible to directly examine and compare the thymopoietic properties of distinct types of epithelial microenvironments in the thymus as a result of the activities of different Foxn1/4 family members.

Foxn1 and its paralog Foxn4 comprise a distinct two-member family of vertebrate wing-helix transcription factors that recognize a unique (G+C)-rich DNA target sequence, distinguished by the core tetranucleotide sequence, 5-ACGC (18, 19). This sequence is also recognized by the DNA binding domain (fig. S1A) encoded by the Foxn4 gene of amphioxus (19), indicating that the evolutionary conservation of the protein sequence of the DNA binding domains encoded by this gene family (4) is mirrored in identical target recognition sequences. In addition to the centrally located DNA binding domain, Foxn1/4 transcription factors exhibit N-terminal domains of variable lengths and acidic transcriptional activation domains (20) in their C-terminal region (fig. S1A); of note, the activation domains are functionally interchangeable, as indicated by the fact that the C terminus of the amphioxus (lancelet) Foxn4 protein can replace that of the mouse Foxn1 protein in in vitro assays of transcriptional activation (21). These observations suggest that the N-terminal regions of Foxn1/4 proteins have been important targets for evolutionary modifications, possibly related to changes in function.

To determine the thymopoietic properties of different members of the Foxn1/4 transcription factor family in the context of the mouse hematopoietic system, we have developed a generic transgenic replacement strategy. It begins with Foxn1-deficient mice, in which the thymic epithelium is functionally inactive (10); it is important to note that, because immature progenitor cells persist in the organ anlage, a functional thymus can be formed upon reactivation of the Foxn1 function (12). We then introduce a transgenic construct that contains all regulatory sequences of the mouse Foxn1 gene (fig. S1B) that are necessary to direct tissue-specific and orthotopic expression of any complementary DNA (cDNA) to the endogenous Foxn1 expression domains into this Foxn1-deficient background. A previous study using this system demonstrated that the expression of the cognate mouse Foxn1 cDNA rescued the pleiotropic nude phenotype in the thymus and the skin (22), thus functionally validating the replacement strategy in vivo. In the first application of this method, we showed that the mouse Foxn4 transcription factor gene [which is paralogous to mouse Foxn1 (4), although not expressed in the mouse thymic epithelium (22)] is nonetheless capable of supporting a degree of lymphopoiesis in the reconstructed thymi (22). These results encouraged us to extend our studies to Foxn1/4 family members (fig. S1C) identifiable in extant representatives of evolutionarily more ancient chordates, using the phenotypes of mouse Foxn1 and mouse Foxn4 replacements as references.

For centuries, biologists have debated the evolutionary origin of vertebrates (23); the current scenario of chordate taxa suggests that lancelets are the sister group to tunicates and vertebrates (24, 25). This phylogenetic relationship is mirrored in our analysis of the chordate Foxn1/4 gene family, which suggests that an ancestral metazoan Foxn4 gene gave rise to the Foxn1 and Foxn4 genes of vertebrates (Fig. 1 and fig. S2) (4). Although tunicates are considered to be phylogenetically closest to vertebrates (24, 25), we chose the Foxn4 gene of amphioxus for our functional analyses, because tunicate development is substantially secondarily modified (26). Hence, to understand the role of Foxn4 in the emergence of specific aspects of the vertebrate body plan, lancelets appeared to be better proxies than tunicates. Moreover, we had previously shown that the amphioxus Foxn4 gene is expressed in the pharyngeal endoderm, the future site of the thymic epithelium in vertebrates (4). For the present experiments, we focused on one species of lancelets (Branchiostoma lanceolatum). To study vertebrate-specific aspects of the Foxn1/4 gene family, we turned to an extant representative of the most ancient group of jawed vertebrates, cartilaginous fishes. To this end, we identified and isolated the Foxn1 and Foxn4 genes of elephant shark (Callorhinchus milii) for our replacement studies.

Vertebrate Foxn1 and Foxn4 clades recapitulate the known phylogenetic relationships of vertebrates (representatives of mammals, monotremes/marsupials, birds, reptiles, amphibians, and bony and cartilaginous fishes are depicted). The Foxn1 proteins in vertebrates form a monophyletic clade. The Foxn4 proteins are paraphyletic in vertebrates and in tunicates. The Foxn4 proteins in lancelets form the base of the tree. The support from 1000 bootstrap replicates is shown as color-coded branches. The vertebrate Foxn1 and Foxn4 clades are boxed in different colors. The scale is in units of average amino acid substitutions per site.

To reconstruct the functional changes that occurred along the evolutionary trajectory of the thymic microenvironment in vertebrates, we examined the developmental fate of mouse hematopoietic progenitors in the different types of thymic microenvironments. Since the Foxn1-deficient thymic epithelium fails to differentiate and does not support lymphoid development (fig. S3) (69), any lymphopoietic activity in the reconstituted thymic microenvironment must be driven by the expression of the respective Foxn1/4 family member under study. The transgenic thymi were examined both for their lymphopoietic capacity and the characteristics of the epithelial microenvironment. Whereas the hematopoietic compartment was analyzed by cell surface markers and in situ analysis of tissue sections, TECs were additionally characterized by RNA sequencing (RNA-seq). Guided by our previous work (22), we focused our attention on three major phenotypic aspects of the thymi that were reconstituted by the Foxn1/4 gene family members of amphioxus and shark. With regards to the composition of the thymic microenvironment, we specifically addressed the question of whether they are capable of supporting the formation of a distinct medullary area, a key compartment associated with the selection of a self-tolerant T cell repertoire (27). With respect to the lymphopoietic properties of the reconstructed thymi, we paid particular attention to their capacity to support the differentiation of thymocytes throughout the known developmental trajectory (17); moreover, we scored the presence and localization of immature and mature B cells, since our previous studies indicated a propensity of the mouse Foxn4 gene to support B cell poiesis when expressed in the thymic epithelium (22), a capacity that the wild-type mouse thymus lacks.

We first examined the thymopoietic capacity of lancelet Foxn4 (Bl_Foxn4), the sole family member of the Foxn1/4 gene family in the genome of the cephalochordate B. lanceolatum. Lancelets lack an adaptive immune system, although cytological evidence for the presence of lymphocyte-like cells has been reported in some species (28, 29). Bl_Foxn4 exhibits some thymopoietic activity. After replacement of mouse Foxn1 (Mus musculus, hereafter Mm_Foxn1) with Bl_Foxn4, the transgenic thymic microenvironment predominantly harbors CD4+CD8+ double-positive (DP) immature thymocytes (Fig. 2A), although their number amounts to only about 1% of that in wild-type thymi (Fig. 2B). Two other features distinguish the hematopoietic compartment of Bl_Foxn4 thymi from the corresponding wild-type situation. First, very few, if any, single-positive T cells are detectable (Fig. 2A), and second, the CD4CD8 double-negative compartment is much larger than in wild-type mice (Fig. 2A). Notably, the absolute number of CD45+CD4CD8CD19+B220+IgM+CD93 mature B cells present in Bl_Foxn4 thymi (Fig. 2C) is only 10-fold lower than that in wild-type thymi (Fig. 2D), indicating that, relative to a mouse wild-type thymus, the transgenic microenvironment tends to be more favorable for mature B cells than for immature T cells.

(A) Flow cytometric analysis of CD45+ thymocytes, stained for T cell markers; wt (n = 6) and Bl_Foxn4 (n = 7). (B) Absolute numbers of total thymocytes and CD4+CD8+ DP T cells in wt (n = 7) and Bl_Foxn4 transgenic thymi (n = 6); ***P < 0.001; two-tailed t test for both groups. (C) Flow cytometric analysis of CD45+ thymocytes and B cell markers; wt (n = 6) and Bl_Foxn4 (n = 7). (D) Absolute numbers of immature CD45+CD4CD8CD19+IgMCD93+ B cells; P = 0.1058, two-tailed t test; wt (n = 6) and Bl_Foxn4 (n = 7). (E) Ly51 expression and UEA1 binding on EpCAM+CD45 TECs; wt (n = 5) and Bl_Foxn4 (n = 5). (F) Heatmap of differential gene expression patterns of TECs. Genes whose expression is associated with particular TEC subsets are indicated (green, cTEC-like; red, mTEC-like; blue, mature cTEC); analysis based on 18,808 protein-coding genes. Scale refers to the percentage of maximum and minimum values of transcript counts of individual genes. (G) Total numbers of CD45EpCAM+ TECs; wt (n = 5) and Bl_Foxn4 (n = 8). (H and I) T and B cell poietic indices calculated from ratios of means (SDs correspond to propagated errors); ***P < 0.001 two tailed t test.

The unique lymphoid signature in the Bl_Foxn4-reconstituted thymi is the result of a microenvironment characterized by epithelial cysts (fig. S4, A and B), dominated by cells reminiscent of cortical TECs (cTECs), as determined by cell surface phenotype (EpCAM+Ly51+UEA1) (Fig. 2E) and the keratin signature (K8+K5) in tissue sections (fig. S4A); we note a conspicuous lack of medullary TECs (mTECs) both in flow cytometric (EpCAM+Ly51UEA1+) (Fig. 2E) and immunohistological (K8K5+) (fig. S4A) analyses. Accordingly, the epithelium has a global transcriptional profile heavily skewed toward a cTEC signature (high levels of Bmp4, Psmb11, Cxcl12, and Dll4 genes), with low levels of genes typically associated with mTECs (Trpm5 and Aire) (30); of note, the near-complete absence of mature TECs is also reflected in low levels of Mhc2 gene expression (Fig. 2F). In the wild-type thymus, the Aire gene is expressed in the CD80+MHCIIhi subset of mature mTEC and involved in the regulation of promiscuous gene expression that is required for proper negative selection of autoreactive T cells (31).

Overall, this constellation gives rise to a low thymopoietic index (calculated as the ratio of hematopoietic cells to TECs) for immature CD4+CD8+ T cells; the capacity of Bl_Foxn4 thymi to generate these immature T cells is two orders of magnitude lower than that of wild-type thymi, compatible with the immature status of the transgenic TECs. Because immature CD45+CD4CD8CD19+B220+IgMCD93+ (henceforth IgMCD93+) B cells are barely detectable in the transgenic thymi (Fig. 2, G to I), the reduction of B poietic capacity can be less confidently determined but appears to be at least 10-fold. Collectively, mice expressing Bl_Foxn4 instead of Mm_Foxn1 in TECs fail to support proper T cell development in the thymus, lack appreciable B cellgenerative capacity, and are most likely incapable of executing the necessary positive and negative selection steps that normally give rise to a self-tolerant repertoire of mature T cells. As a consequence, transgenic mice rapidly (fig. S4C) succumb to an inflammatory syndrome, chiefly affecting the intestine (fig. S4D), occasionally accompanied by vitiligo (fig. S4E). Nonetheless, our data indicate that Bl_Foxn4 is preadapted to support the early stages of T lymphopoiesis when expressed in the context of a vertebrate immune system, attesting to the strong evolutionary conservation of the Notch signaling pathway in regulating the differentiation of hematopoietic precursors [reviewed in (14)].

To assess the functional changes associated with the emergence of a vertebrate form of the Foxn4 gene, we next examined the lymphopoietic capacity of the Foxn4 gene of an extant representative of an evolutionarily ancient group of jawed vertebrates (Fig. 1 and figs. S1 and S2). When the thymic epithelium expresses the Foxn4 gene of the cartilaginous fish C. milii (Cm_Foxn4) instead of Mm_Foxn1, the lymphoid compartment (Fig. 3, A to E) features a large fraction of IgMCD93+ immature B cells, which amounts to about 10% of all hematopoietic cells. This represents an increase of three orders of magnitude when compared to the fraction of immature B cells among the thymocytes of wild-type mice, which averages about 0.01% of all thymocytes. In absolute terms, the Cm_Foxn4 thymi harbor only about 1% of the hematopoietic cell numbers in wild-type mice. Despite the comparatively low number of T cells in the transgenic thymi, their differentiation appears to proceed normally, as reflected by the presence of DP and single-positive thymocytes and the absence of systemic inflammation. These results suggest that the Cm_Foxn4 thymic microenvironment is conducive to both T and B cell differentiation (Fig. 3, F to H); this type of unusual lymphoid bipotency was previously observed in Mm_Foxn4 transgenic thymi (22).

(A to C) Total numbers of CD45+ hematopoietic cells {wt (n = 20), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 11), and Cm_Foxn1/Cm_Foxn4 double transgenics [Cm_dtg (n = 5)]}, DP T cells [wt (n = 20), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5)], and CD93+ immature B cells [wt (n = 13), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 7), and Cm_dtg (n = 5)] in thymi of mice with the indicated genotypes. (D and E) Representative flow cytometric profiles for mice summarized in (A to C). (D) wt (n = 16), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 9), Cm_dtg (n = 5). (E) wt (n = 13), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 7), and Cm_dtg (n = 5). (F) Total numbers of CD45EpCAM+ TECs; wt (n = 17), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5). (G) T cell poietic indices; wt (n = 20), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5). (H) B cell poietic indices; wt (n = 13), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 7), and Cm_dtg (n = 5). In (A) to (C) and (F) to (H), each data point represents one mouse. ***P < 0.001; one-way ANOVA with Tukeys multiple comparison test in (A) to (C) and (F) to (H).

Compared to the situation in Bl_Foxn4 transgenic mice, the epithelial microenvironment of Cm_Foxn4 thymi exhibits the cell surface phenotype (Fig. 4A) and the keratin (Fig. 4B) and gene (Fig. 4C) expression patterns of a more mature cTEC compartment; this is particularly evident from the much higher expression levels of Prss16 and Ccl25 genes that are elevated to wild-type levels (Fig. 4C).

(A) Representative flow cytometric analyses of Ly51 expression and UEA1 binding on TECs for mice; wt (n = 22), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5). (B) Epithelial microenvironment of reconstructed thymi resolved by keratin 5 (K5) (in green) and K8 (in red) staining; 4-week-old mice. m, medulla; c, cortex. Note the small size of the Cm_Foxn4-reconstructed thymus. (C) Differential gene expression patterns in TEC transcriptomes relative to wild-type mice; Bl_Foxn4 (n = 4), Cm_Foxn4 (n = 4), and wt (n = 3). (D) Localization of Aire+ cells relative to cortical (K8) and medullary (K5) areas. (E) Differential gene expression patterns in TEC transcriptomes of transgenic relative to wild-type mice; wt (n = 3), Mm_Foxn4 (n = 6), and Cm_Foxn4 (n = 4). (F) Localization of B220+ B cells (in green) adjacent to ER-TR7+ mesenchyme (in red). PVS, perivascular space. (G) Differential gene expression patterns in TEC transcriptomes of transgenic relative to wild-type mice; wt (n = 3), Cm_Foxn4 (n = 4), Cm_Foxn1 (n = 3), and Cm_dtg (n = 5). In (C), (E), and (G), each data point represents the average value of at least three mice; all values are significantly different from the wild-type genotype (adjusted P values of <0.05), except those data points falling on 0 Arrows indicate the directions of changes in expression levels between transgenic mice.

Although no histologically distinct medullary region is detectable (Fig. 4B), the expression levels of genes indicative of different subsets of mTECs (Fig. 4C) are higher than in the Bl_Foxn4-driven epithelium. Although the morphology of the transgenic thymus lacks the characteristic zonation of cTECs and mTECs that is typical of the wild-type epithelium, low levels of Aire gene expression are detectable (Fig. 4C). Immunohistological analysis indicates the presence of only few Aire+ TECs, which are observed in the edges of epithelial cellfree regions in the reconstituted thymus, in contrast to the obvious Aire+ mTEC clusters in the wild-type thymus (Fig. 4D).

In line with the remarkable sequence conservation of vertebrate Foxn4 proteins (figs. S1, S2, and S5), the structures of the thymic microenvironments driven by Cm_Foxn4 (Fig. 4B) and Mm_Foxn4 (32) genes are essentially identical, despite 500 million years (Ma) of independent evolution. However, compared to the situation in Cm_Foxn4 transgenic mice, Mm_Foxn4-driven epithelia express lower levels of Prss16 and Aire, suggesting that with evolutionary progression, Foxn4 genes have gradually lost the capacity to support maturation of both cTECs and mTECs (Fig. 4E).

Previously, we have shown that the B cells generated in the Mm_Foxn4 thymus are preferentially located in close proximity to the vasculature, i.e., in the mesenchymal perivascular space (22). This finding highlighted the presence of anatomically distinct niches supporting the development of the two principal lymphocyte lineages in a primordial thymus; T cells differentiate in an epithelial environment, whereas B cells differentiate in a mesenchymal niche, similar to the situation in the sinusoidal environment of the bone marrow (33). In notable similarity, B cells in the Cm_Foxn4-expressing thymi are again found in the perivascular space (Fig. 4F), indicating that Foxn4 proteins favor the formation of anatomically separated domains specialized in either T or B cell differentiation.

Next, we tested the thymopoietic capacity of a Foxn1 gene that is suggested by phylogenetic analysis to have emerged from the ancestral vertebrate Foxn1/4-like gene, following a gene duplication event at the base of vertebrate evolution (Fig. 1 and figs. S1 and S2). With respect to mammalian Foxn1 genes, the elephant shark Cm_Foxn1 gene is the evolutionarily most distant form of a Foxn1-like gene of jawed vertebrates examined here (Fig. 1 and figs. S1, S2, and S6). Despite more than 400 Ma of independent evolution, the lymphopoietic capacity of shark Cm_Foxn1 is remarkably similar to that of mouse Mm_Foxn1. The total numbers of CD4+CD8+ immature thymocytes in Cm_Foxn1-expressing thymi approach those observed in mouse wild-type thymi; moreover, differentiation into CD4+ and CD8+ single-positive T cells occurs; as a result, the relative proportions of immature and mature thymocytes are identical to wild-type thymi (Fig. 3, B and D). With respect to intrathymic B cell development, we find that the absolute numbers of immature B cells are about 10-fold lower in the Cm_Foxn1 transgenic thymi than in the corresponding Cm_Foxn4 reconstitution (Fig. 3, C and E); however, they are slightly increased when compared to the wild-type mouse thymus.

As expected from the near-normal hematopoietic compartment in Cm_Foxn1 thymi, their microenvironment also resembles a wild-type mouse thymus. This is evident from the cytometric profiles of Ly51 and UEA1 markers (Fig. 4A) and the anatomical segregation of K5+K8 (medullary) and K5K8+ (cortical) areas in tissue sections (Fig. 4B); except for a somewhat smaller overall size, the histology of the Cm_Foxn1 thymus is essentially identical to the wild-type form. As expected, Aire+ TECs are present in the distinct medullary areas, sparing the thymic cortex (Fig. 4E). The expression patterns of signature genes in Cm_Foxn1 TECs indicate the remarkable similarity to wild-type TECs in the mouse thymus (Fig. 4G). However, subtle differences in the gene expression profiles exist; the increased levels of Prss16 suggest the presence of a bias toward mature cTECs at the expense of certain mTEC populations, for instance, those expressing Trpm5 (Fig. 4G). Nonetheless, our observations suggest that the T cellbiased lymphopoietic properties of Foxn1-like genes, as exemplified by the mammalian thymus, had already emerged in the ancestor of jawed vertebrates and were maintained throughout subsequent evolution.

With respect to the reconstructions described above, it is important to note that they represent an artificial disentanglement of Cm_Foxn1 and Cm_Foxn4 functions, since in the thymus of cartilaginous fish, Foxn1 and Foxn4 paralogs are coexpressed (fig. S7) (22). However, the expression patterns of the two genes are not completely identical; whereas they are both expressed in the shark retina, the expression of Foxn4 in distinct cell clusters in the spinal cord is unique (fig. S7). To mimic the physiological coexpression of both genes in TECs of cartilaginous fish, we generated Cm_Foxn1/Cm_Foxn4 double-transgenic mice (hereafter Cm_dtg). When compared to the number of thymocytes in Cm_Foxn4 and Cm_Foxn1 single-transgenic mice, this constellation of coexpression results in an intermediate number of T cells, although the cellularity is closer to the situation in Cm_Foxn1 thymi (Fig. 3A). Unexpectedly, coexpression of Cm_Foxn1 and Cm_Foxn4 leads to markedly increased numbers of immature B cells, almost two orders of magnitude more than in the Cm_Foxn1 single transgenic (Fig. 3, C and E). The higher capacity for B cell development in the double-transgenic thymus does not affect their anatomical localization; B cells still reside in close proximity to the vasculature, as seen in thymi driven by the expression of Cm_Foxn4 alone (Fig. 4F). The TECs of the bipotent lymphoid organ in double-transgenic mice exhibit a phenotype intermediate between the single transgenics, as reflected in their cell surface phenotype (Fig. 4A); cTECs expressing high levels of Ly51 still predominate, although UEA1+ mTECs make up a quarter of the TEC compartment. The keratin expression pattern indicates that the medullary and cortical regions are not as precisely demarcated as in the Cm_Foxn1 single transgenic (Fig. 4B); however, the medulla contains a large number of Aire+ cells (Fig. 4E), in line with the gene expression pattern (Fig. 4G). In terms of lymphopoietic capacity, the double-transgenic TECs are distinguished by a markedly increased capacity for B cell development (about 10 times higher than that of the Cm_Foxn4 thymus), whereas the capacity for immature T cells is only slightly reduced compared to the Cm_Foxn1 single-transgenic thymus (Fig. 3, G and H). The unique morphological and functional characteristics of the double-transgenic thymus are reflected in an intermediate gene expression pattern, demonstrating that the combination of signature genes selected here faithfully report the thymopoietic characteristics of TECs. Collectively, our results indicate that the coexpression of Cm_Foxn1 and Cm_Foxn4 in the same TECs recapitulates the observed bipotent nature of lymphopoiesis in the thymi of extant cartilaginous fishes (34), providing an important validation of the biological relevance of the present reconstruction strategy.

Whereas Foxn4 proteins are well conserved during the course of vertebrate evolution (fig. S5), Foxn1 proteins are much more variable (Fig. 1 and fig. S6) in particular, with respect to the lengths and amino acid sequence compositions of their N-terminal domains. When viewed through the lens of the mouse proteins, the sequences encoded in coding exons 2 and 3 of mouse Foxn4 are replaced by unrelated sequences in a single exon (coding exon 2) of mouse Foxn1 (fig. S8). This observation suggested that, after gene duplication, the evolution of protein domain(s) in Foxn1 proteins was associated with increasing lymphoid selectivity, from bipotency of Foxn4 to unipotency of Foxn1. We set out to test this hypothesis by creating a chimeric protein, Foxn1*4, in which the sequence encoded by coding exon 2 of the mouse Foxn1 gene was replaced by coding exons 2 and 3 of mouse Foxn4 (Fig. 5A and fig. S8). The overall cellularity in the thymus of Mm_Foxn1*4 transgenic mice was reduced to about 20% of wild-type numbers, but the differentiation of T cells occurred normally, as revealed by the presence of normal percentages of CD4+ and CD8+ single-positive cells (Fig. 5, B and C). However, the CD4CD8 double-negative compartment was increased in the Mm_Foxn1*4 transgenic thymi, likely caused by an increase in the number of B cells (Fig. 5D). Expression of the Mm_Foxn1*4 chimeric protein led to a 10-fold increase of B cell poietic capacity, without appreciable loss of T cell poietic capacity, when compared to Mm_Foxn1 (Fig. 5, E and F).

(A) Schematic representation of the N-terminal domains of mouse Foxn4, Foxn1, and the Foxn1*4 chimera; boxes correspond to exons, and colored lines correspond to conserved amino acid residues in Foxn4 and Foxn1 clades (see figs. S2 and S8 for details). The > sign denotes the DNA binding and activation domains, which are not shown here. (B) Intermediate cellularity of Foxn1*4 thymi (***P < 0.001; two-tailed t test); wt (n = 5), Mm_Foxn4 (n = 6), and Foxn1*4 (n = 13). (C) Enlarged CD4CD8 DN compartment in Foxn1*4 thymi (P < 0.001; two-tailed t test); wt (n = 5) and Foxn1*4 (n = 13). (D) Moderately increased numbers of IgMCD93+ immature B cells (P = 0.293; two-tailed t test, compared to wt); wt (n = 3) and Foxn1*4 (n = 7). (E) Foxn1*4 supports intrathymic T cell development (P = 0.6654; two-tailed t test, compared to wt); wt (n = 5) and Foxn1*4 (n = 13). (F) Increased B cell development (*P = 0.0335; two-tailed t test, compared to wt); wt (n = 8), Mm_Foxn4 (n = 7), and Foxn1*4 (n = 13). In (E) and (F), data for Mm_Foxn4 transgenics (shaded area) are taken from (22). (G) Flow cytometric analyses of Ly51 expression and UEA1 binding of EpCAM+CD45 TECs; wt (n = 5) and Foxn1*4 (n = 13). (H) Epithelial microenvironment of reconstructed thymi resolved by keratin 5 (K5) (in green) and K8 (in red) staining; 4-week-old mice. (I) Localization of B220+ B cells (in green) adjacent to ER-TR7+ mesenchyme (in red); inset shows a higher magnification of the indicated region highlighting the perivascular space. (J and K) Differential gene expression patterns in TECs; see legend in Fig. 4 for details; wt (n = 3), Mm_Foxn4 (n = 6), Foxn1*4 (n = 3), and Cm_dtg (n = 5). In (B), (E), and (F), each data point represents one mouse.

Cell surface markers (Fig. 5G) of TECs in the Mm_Foxn1*4 thymus are reminiscent of the reconstructed thymus in Cm_dtg mice (Fig. 4A). This is evident from a 10-fold greater fraction of Ly51+UEA1 cTECs in the transgenic thymus, with a corresponding reduction of Ly51UEA1+ mTECs. This conclusion is supported by the pattern of K5 and K8 keratin expression and the histological features of cortical and medullary areas (Fig. 5H). Notably, as is the case in Cm_dtg transgenic thymi (Fig. 4F), B cells are situated in the perivascular space of the thymus of Mm_Foxn1*4-transgenic mice (Fig. 5I). Collectively, this finding demonstrates that the sequences in the evolutionarily dynamic N-terminal domains of Foxn1 and Foxn4 proteins have important roles in controlling the extent of B cell development in the thymic microenvironment. In further support of this conclusion, we find that the lancelet Foxn4 protein [which does not support B cell development (Fig. 2, C, D, and I)] lacks most of the conserved amino acid sequence signature of the vertebrate Foxn4 family in this domain (fig. S2). Last, as expected from flow cytometric profiles of TECs and the distinct histological features seen in the tissue sections, the gene expression signature of Foxn1*4-expressing TECs suggests a bias toward the cTEC lineage at the expense of mature mTECs (Fig. 5J), much like the situation in Cm_dtg mice (Fig. 5K).

Our results provide a previously unattainable possibility to compare the transcriptional profiles of thymic microenvironments established by the activity of the cephalochordate Bl_Foxn4, the combinatorial activities of shark Cm_Foxn1 and Cm_Foxn4, and the activity of the mammalian Mm_Foxn1 genes. On the basis of the expression levels of selected TEC signature genes, a clear evolutionary trend becomes apparent: A gradual decrease in expression of cTEC-like genes is accompanied by an increase in expression of the genes that are characteristic of the mTEC compartment (Fig. 6A). This genetic constellation is associated with the more than 100-fold increase in overall T lymphopoietic capacity when comparing the thymic microenvironment established by the activities of the amphioxus Bl_Foxn4 and the mammalian Mm_Foxn1 genes. At the same time, appreciable B lymphopoietic capacity appears to have been a transient phenomenon, absent in Bl_Foxn4-driven and Mm_Foxn1-driven thymic microenvironments but present in the unique Foxn1 and Foxn4 coexpressing microenvironments characteristic of cartilaginous and teleost fishes (22). Hence, we became interested to determine the potential mechanistic basis of the bipotent microenvironment. Our previous experiments (22) suggested that the ratio of Dll4 and Il7 expression levels in the thymic microenvironment is an important determinant of its lymphopoietic properties, with respect to the balance between T and B cell poiesis. The Notch1/Dll4 signaling pathway is essential for the initiation of T cell development in the thymus (5, 15, 16, 35), whereas Il7 functions as a general lymphopoietic growth factor (36). The Dll4 gene is known to be a direct target of the Foxn1 (5, 37) and Foxn4 (4, 38) transcription factors, whereas the expression of Il7 is independent of Foxn1 activity (39). In support of the latter conclusion, we observed similar expression levels of the Il7 gene in all reconstituted TEC compartments examined here. Hence, since the denominator in the Dll4/Il7 ratio is a constant, it follows that the Foxn1/4-dependent expression levels of Dll4 determine the particular type of lymphopoietic activity in the thymus. In the reconstructions with Cm_Foxn1 and Cm_Foxn4 genes, we observed that, compared to the single transgenics, the Cm_dtg thymus had the highest capacity for B cell development (Fig. 6B), in line with the natural bipotency of the thymus of cartilaginous fish (34, 40). The ratio of T cell poiesis and B cell poiesis (here referred to as the T/B index) positively correlated with the ratio of expression levels of Dll4 and Il7 genes (Fig. 6C). In the comparison of Mm_Foxn4, Mm_Foxn1, and Mm_Foxn1*4 transgenics, the same positive relationship between T/B index and Dll4/Il7 ratio holds (Fig. 6, D and E). As compared to the situation in wild-type mice, Dll4 expression is several fold higher in Bl_Foxn4 transgenic microenvironments than it is in Cm_Foxn4-driven TECs, reflecting the T cell bias in the former and the presence of B cell development in the latter. Overall, the Dll4 expression levels vary by about one order of magnitude (Figs. 4, C, E, and G, and 5, J and K). Although it is likely that other factors affect the lymphopoietic properties of the thymic microenvironment, the modulation of Dll4 expression through Foxn1/4 transcription factors emerges as an evolutionarily conserved and functionally relevant mechanism by which the lymphopoietic capacity and the bias for or against B cell development of the thymus could be modulated.

(A) Differential gene expression patterns in TECs; see legend in Fig. 2 for details. (B and D) T and B cell poietic indices; arrows indicate the altered balance between T and B cell generation. (C and E) Ratios of T and B cell indices as a function of the ratios of Dll4 and Il7 expression levels; Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 7), Cm_dtg (n = 5), wt (n = 3), Mm_Foxn4 (n = 6), and Foxn1*4 (n = 7). ***P < 0.001 and *P < 0.05; one-way ANOVA with Tukeys multiple comparison test; SDs correspond to propagated errors. (F) Schematic summarizing gene content and expression characteristics and associated lymphopoietic properties of thymi during vertebrate evolution.

The in vivo reconstitution experiments described here suggest a sequence of events during vertebrate evolution that culminated in the emergence of the T cellbiased thymus. We hypothesize that the primordial vertebrate Foxn4-like gene was expressed in the pharyngeal endoderm in the ancestor common to all vertebrates, much like its ortholog in lancelets (4). After the emergence of lymphocytes, which may have had their evolutionary origin in lymphocyte-like cells of tunicates (41), the Foxn4-expressing patch of pharyngeal endoderm cells may have supported the development of T-like cells; we further propose that this primordial type of lymphopoietic activity initially supported their development only up to the stage where their germline-encoded antigen receptors were expressed (Fig. 6F). Our previous studies demonstrated that the expression of the Notch ligand Dll4 and the chemokine Cxcl12 in Foxn1-deficient TECs suffices to support T cell differentiation up to the CD4+CD8+ DP stage (5), albeit at a much lower efficiency than we observe here in Bl_Foxn4 reconstitutions. Nonetheless, these results collectively suggest that a small number of effector molecules suffice to jump-start the formation of a lymphopoietic environment. We consider it likely that at this particular stage of immune system evolution, the facility of somatic recombination of antigen receptor genes and an associated quality control mechanism(s) mitigating any potential autoreactivity was not yet established (42). Nonetheless, lymphocytes in the vertebrate ancestors immune system may have expressed different kinds of germline-encoded antigen-specific receptors, analogous to pattern recognition receptors; variegated expression of these sensory modules would have afforded early vertebrates with the capability of immune responses and memory functions through clonal proliferation, akin to natural killer cells in mammals (43). Collectively, the phenotype of the Bl_Foxn4 epithelium defines a previously unidentified checkpoint during TEC differentiation, which marks the support of T cell development up to the CD4+CD8+ DP stage.

The emergence of a typical vertebrate-like Foxn4 gene (here exemplified by Cm_Foxn4) heralds another critical transition point in the immune systems of early vertebrates, as it established an environment supporting the development of the two principal lymphocyte lineages. Within the epithelial TEC compartment, it fostered the further development of T cells to reach the single-positive stage, albeit at low efficiency, and at the interface between epithelial and mesenchymal components of the microenvironment, it established conditions conducive to B cell development, as indicated by the presence of substantial numbers of immature B cells in and around the perivascular space. A comparison of protein sequences suggests that changes in the N-terminal domain of Bl_Foxn4 facilitated this process. Since the predicted Foxn4 protein of tunicates assumes an intermediate position in the phylogeny of chordate Foxn1/4 proteins (Fig. 1), it will be of interest to examine its capacity to support the development of B cells.

After the emergence of the Foxn1 gene in a primordial vertebrate, as a result of a gene duplication event, coexpression in the pharyngeal epithelium of the paralogous Foxn4 and Foxn1 genes was maintained; this coexpression signature still persists in extant cartilaginous (fig. S7) (4) and bony fishes (22, 44). Protein sequence comparisons indicate that the emergence of the Foxn1 paralog was accompanied by a radical modification of the amino acid sequence composition of the N terminus. This finding strongly supports the notion that exon replacement event(s) accompanied the emergence of the first Foxn1 genes that exchanged two exons of the Foxn4 gene by a single exon of substantially different sequence. In the Cm_Foxn1 protein, the N terminus is much shorter than it is in the living representatives of evolutionarily more recent taxa, such as the mouse Foxn1 protein, suggesting that structural features of this domain rather than primary amino acid sequence similarities underlie the equivalent functionalities of the Cm_Foxn1 and Mm_Foxn1 proteins.

Because the overall number of TECs present in the thymic lobes was essentially invariant in all reconstructed thymi and similar in magnitude to that in the wild-type mouse thymus, the main difference between Bl_Foxn4 and the vertebrate versions of Foxn1/4 proteins is the much lower lymphopoietic capacity of the former. This observation indicates that the key functions of the Foxn1/4 proteins are to qualitatively alter epithelial cell phenotypes rather than acting to simply quantitatively expand the epithelial compartment via proliferation. Our results indicate that the increase in lymphopoietic capacity occurred in a stepwise fashion, from Bl_Foxn4 to Cm_Foxn4 to CmFoxn1. In addition, the transition from Foxn4 to Foxn1 was accompanied by an increased size of the mTEC compartment, associated with a larger proportion of mature single-positive T cells among thymocytes. In the embryonic mouse thymus, cTECs develop earlier than mTECs (45, 46); this ontogenetic sequence closely resembles the phenotypes we observe in the phylogenetic sequence of Bl_Foxn4 to Cm_Foxn4 to Cm_Foxn1.

Another critical transition in the evolutionary trajectory of Foxn1/4 genes and vertebrate thymopoiesis occurred when cis-regulatory changes led to the loss of Foxn4 expression in TECs. This reorganization of genetic networks is exemplified by the reciprocal expression patterns of Foxn1 and Foxn4 in chicken tissues (fig. S9). As a result, T cell development became entirely dependent on Foxn1 (9, 47). At present, we do not know why a degenerate network structure [note that Foxn4 and Foxn1 are partially redundant in the teleost thymus (22)] was replaced by a nonredundant design. However, since the contribution of the thymus to B cell development was abolished in this process, loss of Foxn4 expression in TECs helped establish the strict anatomical segregation of developing lymphocyte lineages (48). As a result, the thymus was eventually transformed into an organ highly specialized for efficient T cell development (Fig. 6F).

C57BL/6J mice are maintained in the Max Planck Institute of Immunobiology and Epigenetics. Foxn1:Bl_Foxn4, Foxn1:Cm_Foxn4, and Foxn1:Cm_Foxn1 transgenic mice were constructed according to standard protocols by cloning a 27,970base pair mouse Foxn1 promoter fragment (GenBank accession number Y12488; nucleotides 5680 to 33,650) upstream of either B. lanceolatum Foxn4 cDNA (GenBank accession number AJ252025.1; nucleotides 1 to 1590), C. milii Foxn4 cDNA (GenBank accession number FJ176202.1, nucleotides 56 to 1615), or a C. milii Foxn1 cDNA (GenBank accession number FJ176201.1; nucleotides 76 to 1584), followed by the bovine growth hormone polyadenylation sequence (32); cDNAs of Cm_Foxn4 and Cm_Foxn1 were synthesized by Eurofins Genomics and cloned into peX-K248 vector; in the Foxn1:Foxn1*4 construct, nucleotides 267 to 728 of M. musculus Foxn1 (GenBank accession number NM_008238.2) were replaced by nucleotides 170 to 445 of M. musculus Foxn4 (GenBank accession number AF323488.1). To generate transgenic mice, constructs were linearized and injected into FVB pronuclei according to standard protocols.

Transgenic mice were subsequently backcrossed to Foxn1-deficient mice (10) on a C57BL/6J background. Mice were kept in the animal facility of the Max Planck Institute of Immunobiology and Epigenetics under specific pathogenfree conditions. All animal experiments were performed in accordance with the relevant guidelines and regulations, approved by the review committee of the Max Planck Institute of Immunobiology and Epigenetics and the Regierungsprsidium Freiburg, Germany (license AZ 35-9185.81/G-14/57). Transgene expression levels were determined by RNA-seq, comparing transgene-derived BGH_PolyA transcripts to lacZ transcripts, the latter representing the activity of the targeted endogenous Foxn1 locus (10); the ratios of lacZ transcript counts to those emanating from the 5 part of the Foxn1 gene [note that the 5 end of the Mm-Foxn1 sequence is still detectable in the transcriptome of Foxn1/ mice (10)] served as normalization. A negative feedback loop suppresses the endogenous Foxn1 locus activity for transgenes encoding proteins functionally equivalent to Foxn1; despite a 10-fold difference in expression levels, the transgenes are expressed in the range of the endogenous Foxn1 gene in wild-type mice (fig. S10). For identification of transgenes, the following primers were used for genotyping: Foxn1:Mm_Foxn1, SS40 (wild-type allele) + SS35 (wild-type and knockout allele) + JBS003 (knockout allele), Foxn1:Mm_Foxn1*4, XAH388 + SS6; Foxn1:Cm_Foxn1, XAH163 + RM19; Foxn1:Cm_Foxn4, XAH163 + RM22; Foxn1:Bl_Foxn4, JBS465 + JBS466; SS40, 5-CTGTGAACTCAGCCATACTC; SS35, 5-TGCACCAAGCCTCTGCTGGGA; JBS003, 5-TCGCCTTCTTGACGAGTTCT; XAH388, 5-CAGCAACTGATAAGGTCACC; SS6, 5-ACAGAATTCTTCCAGCCATCA; XAH163, 5-GTCCCTAATCCGATGGCTAGCTC; RM19, 5-TATCGCGTGCACGAGTTGTA; RM22, 5-GGTTAAAGTTCATGCGGCCG; JBS465, 5-CCAGCTCCGAAACAGCCTAA; JBS466, 5-GTCCTTTGTCGTCTGGTCGT.

Thymus organs were fixed for 120 min in 4% paraformaldehyde, washed with phosphate-buffered saline (PBS), and incubated in 20% sucrose overnight before mounting and snap-freezing in optimal cutting temperature (OCT) embedding compound. Tissue sections (8 m) were cut using a cryostat and mounted onto precoated slides (Superfrost plus, Thermo Fisher Scientific). Slides were dried, followed by a 30-min blocking step using mouse immunoglobulin G (IgG) at 1:50 diluted in PBS + 0.5% bovine serum albumin (BSA) + 0.2% Tween. K5 K8 staining was performed with rabbit anti-K5 antibody (Ab) (PRB-160P, Covance) at 1:500 and rat anti-K8 Ab (Troma1, in-house) at 1:200. As secondary Ab, goat anti-rabbit Alexa Fluor 488 (A11008, Thermo Fisher Scientific) at 1:500 and donkey anti-rat Cy3 (AB_2340668, Jackson ImmunoResearch) at 1:500 were used. For ER-TR7 B220 staining, the rat antiER-TR7 Alexa Fluor 647 Ab (sc-73355 AF647, Santa Cruz Biotechnology) at 1:50 and rat anti-B220 Alexa Fluor 488 Ab (RA3-6B2, eBioscience) at 1:200 were used. Sections were mounted with Fluoromount G before analysis (Apotome, Zeiss). For combined K5/K8/Aire staining, sections were dried, blocked, and stained with unlabeled primary Abs as above and then stained with the secondary Abs donkey anti-rabbit IgG Cy3 (Jackson ImmunoResearch 711-165-152) and mouse anti-rat light chain Alexa Fluor 647 (MAR 18.5.28, purified and labeled in-house). Sections were then blocked with rat IgG and subsequently stained with rat anti-mouse Aire Alexa Fluor 488 Ab (5H12, eBioscience 14-5934-82) at 1:200.

RNA in situ hybridization was carried out essentially as described (22) using the following probes: Gallus gallus Foxn1, nucleotides 1371 to 2557 in GenBank accession number XM_415816.6; G. gallus Foxn4, nucleotides 559 to 1779 in GenBank accession number NM_001083359.1; Scyliorhinus canicula Foxn1, nucleotides 37 to 346 in GenBank accession number FJ187748; and S. canicula Foxn4, nucleotides 1 to 422 combined from GenBank accession numbers Y11538, Y11539, and Y11540, respectively.

To generate single-cell suspensions for analytical and preparative flow cytometry of TECs, the procedures in (49) were followed. Note that the enzymatic cocktail required to liberate TECs destroys the extracellular domains of CD4 and CD8 surface markers (but not that of the CD45 molecule); hence, when analysis of thymocyte subsets was desired, thymocyte suspensions were prepared in parallel by mechanical liberation, best achieved by gently pressing thymic lobes through 40-m sieves. Cell surface staining [anti-CD45 (30-F11), conjugated with phycoerythrin (PE) Cy7 (BioLegend); anti-EpCAM (G8.8), conjugated with allophycocyanin (APC; BioLegend); anti-Ly51 (BP-1) (6C3), conjugated with PE (eBioscience); UEA1, conjugated with fluorescein isothiocyanate (FITC; Vector Biosciences); anti-CD4 (GK1.5), conjugated with FITC (BioLegend); anti-CD8a (53-6.7), conjugated with APC (eBioscience); anti-CD19 (eBio1D3), conjugated with PerCPCy5.5 or PeCy7 (eBioscence); anti-B220 (CD45R) (RA3-6B2), conjugated with biotin (eBioscience); anti-IgM (II/4.1), conjugated with PE (eBioscience), anti-CD93 (C1qRp) (AA4.1), conjugated with APC (eBioscience); streptavidin conjugated with eFluor 450 or FITC (eBioscience)] was performed at 4C in PBS supplemented with 0.5% BSA and 0.02% NaN3. Because of their small size in Bl_Foxn4 mice, the numbers of TECs and hematopoietic cells in thymi were determined independently; hence, B and T poietic indices were calculated from mean values with error propagation.

Single-cell suspensions were prepared by TEC digest as described above. CD45EpCAM+ cells [negative enrichment using anti-CD45 magnetic-activated cell sorting (MACS) beads and antiTer-119 MACS beads, Miltenyi Biotec] were sorted directly into TRI reagent (T9424, Sigma-Aldrich). RNA isolation was performed according to standard protocols. Libraries were prepared using the Ultra RNA Library Prep Kit (Illumina). Samples were run on HiSeq2500 and sequenced to a depth of > 60 106 to 100 106 reads per sample.

The relevant Foxn1 and Foxn4 amino acid sequences can be found under the following GenBank accession numbers: Foxn1: Rhincodon typus (XM_020525471); C. milii (XM_007896499); Amblyraja radiata (XM_033046380); Erpetoichthys calabaricus (XM_028808185); Acipenser ruthenus (XM_034049625); Lepisosteus oculatus (XM_015367325); Danio rerio (XM_009291615); Microcaecilia unicolor (XM_030186024.1); Nanorana parkeri (XM_018555008.1); Xenopus laevis (XM_018248776.1); Xenopus tropicalis (XM_018091796); Podarcis muralis (XM_028708697); Geotrypetes seraphini (XM_033921305); Anolis carolinensis (XM_016997340); G. gallus (XM_415816); Corvus cornix cornix (XM_019287554); Monodelphis domestica (XM_001375795); Ornithorhynchus anatinus (XM_029082501); M. musculus (NM_008238); Homo sapiens (NM_001369369); Foxn4: B. lanceolatum (AJ252025); Branchiostoma belcheri (XP_019621093); Phallusia mammillata (LR785254); R. typus (XM_020536376); A. radiata (XM_033043787); C. milii (NM_001292643); L. oculatus (XM_006640210); E. calabaricus (XM_028824927); D. rerio (NM_131099); Latimeria chalumnae (XM_014493627); Rhinatrema bivittatum (XM_029571604); X. laevis (BC142562); X. tropicalis (NM_001102862); G. seraphini (XM_033957269); P. muralis (XM_028705153); G. gallus (NM_001083359); C. cornix cornix (XM_020584559); Phascolarctos cinereus (XM_021008024); M. musculus (NM_148935); H. sapiens (NM_213596). The sequence of Foxn4 of Ciona intestinalis has been retrieved from the ENSEMBL database (ENSCING00000017653). Sequences were aligned using multiple sequence comparison by log-expectation (MUSCLE) (50), and the phylogenetic tree was reconstructed using the neighbor-joining method implemented in the BioNJ software (51) with 1000 bootstrap replicates and the Jones-Taylor-Thornton (JTT) substitution model (52). Both programs are available on the platform phylogeny.fr (53). The resulting phylogeny was visualized using the Interactive Tree of Life platform (54).

Transcriptomes were analyzed on the Galaxy platform using featureCounts (55) followed by DESeq2 analysis (56).

t tests (two-tailed) were used to determine the significance levels of the differences between the means of two independent samples, considering equal or unequal variances as determined by the F test. For multiple tests, the conservative Bonferroni correction was applied or as indicated, using one-way analysis of variance (ANOVA) with Tukeys multiple comparison test.

Excerpt from:
Retracing the evolutionary emergence of thymopoiesis - Science Advances

Read More...

Exosomes derived from BDNF-expressing 293T attenuate ischemic retinal injury and . – Physician’s Weekly

December 4th, 2020 12:15 am

Retinal ischemia emerges in many ocular diseases and is a leading cause of neuronal death and dysfunction, resulting in irreversible visual impairment. We previously reported that brain-derived neurotrophic factor (BDNF)-expressing human 293T cells could steadily express BDNF and play a protective role in ARPE-19 cells, a human retinal epithelial cell line. Thus, we hypothesized that exosomes might be essential in the interaction between BDNF-expressing 293T cells and recipient cells. The study investigated whether exosomes derived from BDNF-expressing 293T cells (293T-Exo) can be internalized by ischemic retinal cells and exert neuroprotective roles. The results demonstrated that 293T-Exo significantly attenuated the loss of cell proliferation and cell death in R28 cells in response to oxygen-glucose deprivation treatment. Mechanistic studies revealed that the endocytosis of 293T-Exo by R28 cells displayed dose- and temperature-dependent patterns and may be mediated by the caveolar endocytic pathway via the integrin receptor. In the retinal ischemia rat model, the administration of 293T-Exo into the vitreous humor of ischemic eyes reduced apoptosis in the retina. Furthermore, 293T-Exo was mainly taken up by retinal neurons and retinal ganglion cells. Together, the results demonstrated that 293T-Exo has a neuroprotective effect in retinal ischemia and has therapeutic potential for retinal disorders.

PubMed

Read the rest here:
Exosomes derived from BDNF-expressing 293T attenuate ischemic retinal injury and . - Physician's Weekly

Read More...

Opthalmic Drugs Growth Investigation Reveals Enhanced Growth during the forecast Period, 2020 2025 – The Cloud Tribune

December 4th, 2020 12:15 am

The report highlights the key growth drivers and trends that will contribute to the growth momentum of the market. The report provides an incisive analysis of the growth dynamics and quantitative assessment of the revenue potential in various regions and across key products, application/end-use industry, and technology segments. It provides a comprehensive insight into the shares and sizes of the various segments in each year of the forecast period. The assessment of the growth dynamics in the Opthalmic Drugs Market cover the year-over-year growth of key geographies, and the incremental opportunities in numerous key countries.

The authors of the report have made a rigorous assessment of various industry-specific trends and analysis of the macroeconomic environment prevailing in various regions around the world.

Get Sample Copy of this Report https://www.factmr.com/connectus/sample?flag=S&rep_id=4799

A comprehensive evaluation of recent investments by top players in the Opthalmic Drugs market helps identify major research and development initiatives in the key regional markets. The various assessments on the competitive landscape focus on the intensity of competition, entry barriers, PESTLE analysis, and key winning imperatives. The readers can further find pertinent information on the recent market developments such as divestments, entry of players from different industries, licensing deals, and long-term partnerships to consolidate shares by top players.

Key companies profiled in the study are:

The study further identifies major manufacturing trends, technologies that will be commercialized, and

The detailed market estimations cover the following:

How will be insights and market estimations provided in the Fact.MR report on the Opthalmic Drugs market make a difference:

A Customization of this Report is Available upon Request https://www.factmr.com/connectus/sample?flag=RM&rep_id=4799

The Opthalmic Drugs market is segmented on the basis of geography into:

On the basis of product types,

The study identifies the following application areas that will be at the center of investments are:

About Fact.MR

Fact.MR is a fast-growing market research firm that offers the most comprehensive suite of syndicated and customized market research reports. We believe transformative intelligence can educate and inspire businesses to make smarter decisions. We know the limitations of the one-size-fits-all approach; thats why we publish multi-industry global, regional, and country-specific research reports.

Contact:

Unit No: AU-01-H Gold Tower (AU), Plot No: JLT-PH1-I3A,

Jumeirah Lakes Towers, Dubai, United Arab Emirates

MARKET ACCESS DMCC Initiative

Email: sales@factmr.com

Web: https://www.factmr.com/

Excerpt from:
Opthalmic Drugs Growth Investigation Reveals Enhanced Growth during the forecast Period, 2020 2025 - The Cloud Tribune

Read More...

Opthalmic Therapeutics Market Size, Share & Trends Analysis Report By Product Types, And Applications Forecast To 2026 – The Market Feed

December 4th, 2020 12:15 am

The Global Opthalmic Therapeutics Market analysis report published on dataintelo is a detailed study of market size, share and dynamics covered in XX pages and is an illustrative sample demonstrating market trends. This is a latest report, covering the current COVID-19 impact on the market. The pandemic of Coronavirus (COVID-19) has affected every aspect of life globally. This has brought along several changes in market conditions. The rapidly changing market scenario and initial and future assessment of the impact is covered in the report. It covers the entire market with an in-depth study on revenue growth and profitability. The report also delivers on key players along with strategic standpoint pertaining to price and promotion.

Get FREE Exclusive PDF Sample Copy of This Report: https://dataintelo.com/request-sample/?reportId=44733

The Global Opthalmic Therapeutics Market report entails a comprehensive database on future market estimation based on historical data analysis. It enables the clients with quantified data for current market perusal. It is a professional and a detailed report focusing on primary and secondary drivers, market share, leading segments and regional analysis. Listed out are key players, major collaborations, merger & acquisitions along with upcoming and trending innovation. Business policies are reviewed from the techno-commercial perspective demonstrating better results. The report contains granular information & analysis pertaining to the Global Opthalmic Therapeutics Market size, share, growth, trends, segment and forecasts from 2020-2026.

With an all-round approach for data accumulation, the market scenarios comprise major players, cost and pricing operating in the specific geography/ies. Statistical surveying used are SWOT analysis, PESTLE analysis, predictive analysis, and real-time analytics. Graphs are clearly used to support the data format for clear understanding of facts and figures.

Customize Report and Inquiry for The Opthalmic Therapeutics Market Report: https://dataintelo.com/enquiry-before-buying/?reportId=44733

Get in touch with our sales team, who will guarantee you to get a report that suits your necessities.

Primary research, interviews, news sources and information booths have made the report precise having valuable data. Secondary research techniques add more in clear and concise understanding with regards to placing of data in the report.

The report segments the Global Opthalmic Therapeutics Market as:Global Opthalmic Therapeutics Market Size & Share, by Regions

Global Opthalmic Therapeutics Market Size & Share, by ProductsProduct Type IProduct Type IIProduct Type III

Global Opthalmic Therapeutics Market Size & Share, ApplicationsApplication IApplication IIApplication III

Key PlayersActavisBayerF. Hoffmann-La RocheMerckNovartisPfizer

Avail the Discount on this Report @ https://dataintelo.com/ask-for-discount/?reportId=44733

Dataintelo offers attractive discounts on customization of reports as per your need. This report can be personalized to meet your requirements. Get in touch with our sales team, who will guarantee you to get a report that suits your necessities.

About DataIntelo:DATAINTELO has set its benchmark in the market research industry by providing syndicated and customized research report to the clients. The database of the company is updated on a daily basis to prompt the clients with the latest trends and in-depth analysis of the industry. Our pool of database contains various industry verticals that include: IT & Telecom, Food Beverage, Automotive, Healthcare, Chemicals and Energy, Consumer foods, Food and beverages, and many more. Each and every report goes through the proper research methodology, validated from the professionals and analysts to ensure the eminent quality reports.

Contact Info:

Name: Alex Mathews

Address: 500 East E Street, Ontario,

CA 91764, United States.

Phone No: USA: +1 909 545 6473

Email:[emailprotected]

Website:https://dataintelo.com

Read more here:
Opthalmic Therapeutics Market Size, Share & Trends Analysis Report By Product Types, And Applications Forecast To 2026 - The Market Feed

Read More...

Opthalmic Therapeutic Market Research Report 2020 with Manufacturing Process Analysis and Market Concentration Rate till 2026 – The Market Feed

December 4th, 2020 12:15 am

The Opthalmic Therapeutic Market grew in 2019, as compared to 2018, according to our report, Opthalmic Therapeutic Market is likely to have subdued growth in 2020 due to weak demand on account of reduced industry spending post Covid-19 outbreak. Further, Opthalmic Therapeutic Market will begin picking up momentum gradually from 2021 onwards and grow at a healthy CAGR between 2021-2025

Deep analysis about market status (2016-2019), competition pattern, advantages and disadvantages of products, industry development trends (2019-2025), regional industrial layout characteristics and macroeconomic policies, industrial policy has also been included. From raw materials to downstream buyers of this industry have been analysed scientifically. This report will help you to establish comprehensive overview of the Opthalmic Therapeutic Market

Get a Sample Copy of the Report at: https://i2iresearch.com/report/global-opthalmic-therapeutic-market-2020-market-size-share-growth-trends-forecast-2025/#download-sample

The Opthalmic Therapeutic Market is analysed based on product types, major applications and key players

Key product type:OralInjectionExternal Application

Key applications:HospitalsClinicsHome Care SettingsOther

Key players or companies covered are:PfizerNovartisGlaxoSmithKlineAlconMerckRocheBayerActavisBoehringer IngelheimR-Tech UenoValeant PharmaceuticalsRegeneron PharmaceuticalsAcadia PharmaceuticalsAllerganFalcon PharmaceuticalsOtsuka PharmaceuticalSenju PharmaceuticalOPKO HealthLexicon Pharmaceuticals

The report provides analysis & data at a regional level (North America, Europe, Asia Pacific, Middle East & Africa , Rest of the world) & Country level (13 key countries The U.S, Canada, Germany, France, UK, Italy, China, Japan, India, Middle East, Africa, South America)

Inquire or share your questions, if any: https://i2iresearch.com/report/global-opthalmic-therapeutic-market-2020-market-size-share-growth-trends-forecast-2025/

Key questions answered in the report:1. What is the current size of the Opthalmic Therapeutic Market, at a global, regional & country level?2. How is the market segmented, who are the key end user segments?3. What are the key drivers, challenges & trends that is likely to impact businesses in the Opthalmic Therapeutic Market?4. What is the likely market forecast & how will be Opthalmic Therapeutic Market impacted?5. What is the competitive landscape, who are the key players?6. What are some of the recent M&A, PE / VC deals that have happened in the Opthalmic Therapeutic Market?

The report also analysis the impact of COVID 19 based on a scenario-based modelling. This provides a clear view of how has COVID impacted the growth cycle & when is the likely recovery of the industry is expected to pre-covid levels.

Contact us:i2iResearch info to intelligenceLocational Office: *India, *United States, *GermanyEmail: [emailprotected]Toll-free: +1-800-419-8865 | Phone: +91 98801 53667

Read this article:
Opthalmic Therapeutic Market Research Report 2020 with Manufacturing Process Analysis and Market Concentration Rate till 2026 - The Market Feed

Read More...

Opthalmology Pacs Market- Roadmap for Recovery from COVID-19| Industry Report Market Outlook and Analysis Through 2028 – The Haitian-Caribbean News…

December 4th, 2020 12:15 am

The Opthalmology Pacs Market Industry report shields a thorough investigation towards the competitive profile encompassing the market stake along with company outline of the major contributors functioning in the global Opthalmology Pacs market. The market report offers a comprehensive summarization of product description, product type, technological development as well manufacturing analysis including cost, income, and gross analysis. The market comprises of past data related to growth rate, market price, volume and futuristic analysis of the Opthalmology Pacs market.

Moreover, it describes factors that are responsible for influencing for the growth of the Opthalmology Pacs market, demand, and supply as well as the challenges and opportunities tackled by the competitive Opthalmology Pacs market. In addition, the report holds important information for the Opthalmology Pacs market players that enables them to understand the overall market scenario and expand their Opthalmology Pacs Market business stats.

The report covers a number of the players in the Opthalmology Pacs market, including:

Topcon Corporation, IBM corporation, Carl Zeiss Meditec AG, EyePACS, Heidelberg Engineering and more.

Request sample copy of this report at: https://www.adroitmarketresearch.com/contacts/request-sample/608

The Opthalmology Pacs market report delivers screen-shot of major competitors, market tendencies together with the forecast over the next five years, with anticipated growth rates and the major factors impacting and driving growth, the market statistics and Opthalmology Pacs Market Industry information resulting from the various blending of primary and secondary sources.

Global Opthalmology Pacs Market Report tracks the all the significant market occasions. Social occasion of data from different fields and through proper discoveries, the report has firmly anticipated development of the worldwide Opthalmology Pacs market including regions and different section.

Key points considered in the Opthalmology Pacs market report: company profile, production cost structure of market, sales and income analysis of Opthalmology Pacs market, production scrutiny by geological region, Opthalmology Pacs market strategies considering the major aspects related to restraint, opportunities, driving factors, challenges and possible analysis of new Opthalmology Pacs market projects and their investment structure.

Read complete report @ https://www.adroitmarketresearch.com/industry-reports/opthalmology-pacs-market

Global Opthalmology Pacs market is segmented based by type, application and region.

Based on Type, the market has been segmented into:

By End-Use, market is segmented into:

HospitalsAmbulatory Surgical Center (ASCS) & Specialty ClinicsOthersBy Type, market is segmented into:

Standalone PACSIntegrated PACSBy Delivery Model, market is segmented into:

Cloud/ web based modelsOn-premise modelsOthers

The Global Opthalmology Pacs market focuses on the major factors mentioned below:

1.A comprehensive outline of the Opthalmology Pacs market has been analyzed, which involves the valuation of the different organization in the global market.2.Developing trends in various Opthalmology Pacs market segments and geological market.3.Substantial changes in market strategies and Opthalmology Pacs market synopsis.4.Market methodologies and market stakes of major players in the Opthalmology Pacs market.5.Existing and future dimensions of Opthalmology Pacs market on the basis of both cost and volume point of view.6.Estimates of latest Opthalmology Pacs Market Industry trends and advanced development.7.Major references for the new entrants for stability in the global and competitive market.

To describe and classify the market for Opthalmology Pacs market

Delivers complete data associated with the major factors inducing growth of the global Opthalmology Pacs market that includes drivers, restraints, opportunities, and trends. Studies and predicts the market volume and market stake of Opthalmology Pacs market, related to volume and values. The Opthalmology Pacs market report examines the market breakdown and anticipates the market volume related to volume and value, for geographical regions that include growth regions over the globe. Investigates competitive expansion, associated with product introduction, developmental stability and agreement & mergers occurring in the Opthalmology Pacs market. Tactically portrays the competitive players functioning in the Opthalmology Pacs market.

Delivers complete data associated with the major factors inducing growth of the global Opthalmology Pacs market that includes drivers, restraints, opportunities, and trends. Studies and predicts the market volume and market stake of Opthalmology Pacs market, related to volume and values. The Opthalmology Pacs market report examines the market breakdown and anticipates the market volume related to volume and value, for geographical regions that include growth regions over the globe. Investigates competitive expansion, associated with product introduction, developmental stability and agreement & mergers occurring in the Opthalmology Pacs market. Tactically portrays the competitive players functioning in the Opthalmology Pacs market.

In this Opthalmology Pacs market report study, scrutiny of dealers and distributors is specified along with contact information. Also, the Opthalmology Pacs market report includes the manufacturing plants, Opthalmology Pacs Market Industry details of imports and exports, demand and supply chain, their ability, worldwide productivity, and revenue. Lastly, it provides the data related to research findings, Opthalmology Pacs Market data sources, conclusion, and appendix.

For Any Query on the Opthalmology Pacs market: https://www.adroitmarketresearch.com/contacts/enquiry-before-buying/608

About Us :

Contact Us :

Ryan JohnsonAccount Manager Global3131 McKinney Ave Ste 600, Dallas,TX75204, U.S.A.Phone No.: USA: +1 972-362 -8199/ +91 9665341414

View original post here:
Opthalmology Pacs Market- Roadmap for Recovery from COVID-19| Industry Report Market Outlook and Analysis Through 2028 - The Haitian-Caribbean News...

Read More...

Global Opthalmology Pacs Market Share 2020 Industry Size, Types and Application, Regions, Manufacturers and Growth Opportunities by 2025 – Khabar…

December 4th, 2020 12:15 am

The research report titled Global Opthalmology Pacs Market Research Report And Predictive Business Strategy By 2025 and published by Adroit Market Research is an in-depth and dedicated scrutiny of the existing stats of the global Opthalmology Pacs Market entailing the numerous facets pertinent to statistics and growth of the business. The report segregated into diverse sections to simplify the comprehension of the included data and thus, market dynamics. It encompasses all the major competitors and players along with the various features relating to the market players like company profiles, supply chain value, product specifications, market shares, and so on. Also, the report entails the major strategic market developments, comprising R&D activities, collaborations, new product launch, agreements, joint ventures, partnerships, M&A, and presence & expansion extent of these prominent players on the global and regional scale. In addition, it comprises the systematic examination of business strategies for expansion of the prominent Opthalmology Pacs Market players.

Download Free PDF Report Brochure @ https://www.adroitmarketresearch.com/contacts/request-sample/608?utm_source=amr

The Opthalmology Pacs Market report encompasses the general idea of the global Opthalmology Pacs Market including definition, classifications, and applications. Further, it includes the all-inclusive comprehension of several factors such as drivers, constraints, and major micro markets. The report is a wide-ranging source of widespread facts and figures for business strategists as it offers the historical & futuristic data such as demand & supply data, cost, revenue, profit, supply chain value, and so on. Furthermore, it entails the key market features, comprising production, revenue, price, capacity, gross margin, market share, consumption, gross, production rate, demand/supply, cost, capacity utilization rate, export/import, and CAGR (compound annual growth rate). In addition the report encompasses global Opthalmology Pacs Market segmentation on the basis of diverse facets like product/service type, application, technology, end-users, and major geographic regions Latin America, North America, Asia Pacific, Middle & East Africa, and Europe. Apart from this, the researcher market analyst and experts present their outlook or insights of product sales, market share, and value along with the possible opportunities to grow or tap into in these regions.

Some of the Major Market Players Are

Topcon Corporation, IBM corporation, Carl Zeiss Meditec AG, EyePACS, Heidelberg Engineering and more.

Request impact analysis on this market @ https://www.adroitmarketresearch.com/industry-reports/opthalmology-pacs-market?utm_source=amr

Promising Regions & Countries Mentioned In The Opthalmology Pacs Market Report:

North America ( United States)Europe ( Germany, France, UK)Asia-Pacific ( China, Japan, India)Latin America ( Brazil)The Middle East & Africa

The Opthalmology Pacs Market report also entails the vigorous evaluation about the growth plot and all opportunities & risk related to of global Opthalmology Pacs Market during the forecast period. In addition, the report comprises the key events and most recent innovations in the industry together with the prospective trends technological progresses within the global Opthalmology Pacs Market that can impact its expansion graph. Entailing the pivotal data on the markets statistics and dynamics, the report will serve as a valued asset in term of decision-making and guidance for the businesses and companies already active within industry or looking forward to enter into it.

Opthalmology Pacs Market by Category:

By Type:

By End-Use, market is segmented into:

HospitalsAmbulatory Surgical Center (ASCS) & Specialty ClinicsOthersBy Type, market is segmented into:

Standalone PACSIntegrated PACSBy Delivery Model, market is segmented into:

Cloud/ web based modelsOn-premise modelsOthers

The Study Objectives of Opthalmology Pacs Market Report Are:

Examine and study the global Opthalmology Pacs Market sales, value, status (2020) and forecast (2025).Focuses on the key Opthalmology Pacs Market manufacturers, to study the sales, value, market share and development plans in the future.Define, describe and forecast the Opthalmology Pacs Market by type, application, and region.Study the global and key regions market potential and advantage, opportunity and challenge, restraints and risks.Know significant trends and factors driving or inhibiting the Opthalmology Pacs Market growth.Study the opportunities in the market for stakeholders by identifying the high growth segments.Strategically examines each submarket with respect to individual growth trend and their contribution to the Opthalmology Pacs MarketExamine competitive developments such as expansions, agreements, new product launches, and acquisitions in the marketStrategically profile the key players and comprehensively analyses their growth strategies.Request coronavirus impact analysis on sectors and market

Some Major Points in TOC:

Chapter 1. Report OverviewChapter 2. Global Growth TrendsChapter 3. Market Share by Key PlayersChapter 4. Breakdown Data by Type and ApplicationChapter 5. Market by End Users/ApplicationChapter 6. COVID-19 Outbreak: Opthalmology Pacs Market Industry ImpactChapter 7. Opportunity Analysis in Covid-19 CrisisChapter 9. Market Driving Force

Also, Research Report Examines:

1.Competitive companies and manufacturers in global market2.By Product Type, Applications & Growth Factors3.Industry Status and Outlook for Major Applications / End Users / Usage Area4.Thanks for reading this article; you can also get individual chapter wise section or region wise report version like North America, Europe or Asia.

Inquire more about this report @ https://www.adroitmarketresearch.com/contacts/enquiry-before-buying/608?utm_source=amr

About Us :

Contact Us :

Ryan JohnsonAccount Manager Global3131 McKinney Ave Ste 600, Dallas,TX75204, U.S.A.Phone No.: USA: +1 972-362 -8199/ +91 9665341414

See more here:
Global Opthalmology Pacs Market Share 2020 Industry Size, Types and Application, Regions, Manufacturers and Growth Opportunities by 2025 - Khabar...

Read More...

Opthalmology Pacs Market Global Innovations, Competitive Analysis, New Business Developments and Top Companies Global Forecast to 2026 – Cheshire…

December 4th, 2020 12:15 am

Global Opthalmology Pacs Market (2020-2026) status and position of worldwide and key regions, with perspectives of manufacturers, regions, product types and end industries; this report analyses the topmost companies in worldwide and main regions, and splits the Opthalmology Pacs market by product type and applications/end industries. The Opthalmology Pacs market trend research process includes the analysis of different factors affecting the industry, with the government policy, competitive landscape, historical data, market environment, present trends in the market, upcoming technologies, technological innovation, and the technical progress in related industry, and market risks, market barriers, opportunities, and challenges.

Get Sample Copy of this Report: https://www.adroitmarketresearch.com/contacts/request-sample/608

Top Leading Key Players are:

Topcon Corporation, IBM corporation, Carl Zeiss Meditec AG, EyePACS, Heidelberg Engineering and more.

Global Opthalmology Pacs Market 2020 report spotlights major statistics of the current industry state and is a beneficial source of developments and opportunities for individuals and firms interested in the Opthalmology Pacs industry. The report primarily concentrate on the Opthalmology Pacs market growth in productivity, demand, trade and investment with company profiles, specification and product picture. Worldwide Opthalmology Pacs market study predicts revenues for applications across key regions with scope of Opthalmology Pacs market, manufacturing cost structure analysis, and product overview. The industry report analyzes the world Opthalmology Pacs market estimates and forecasts of all the given segments on global as well as regional levels. The study provides historical Opthalmology Pacs market data couples with revenue predictions and forecasts from 2020 till 2026. The report focuses on Opthalmology Pacs market trends, supply chain trends, technical modernization, leading players, key developments, and Opthalmology Pacs future strategies

Enquire for in-depth information before buying this report @ https://www.adroitmarketresearch.com/contacts/enquiry-before-buying/608

Global Opthalmology Pacs market is segmented based by type, application and region.Based on Type, the market has been segmented into:

By End-Use, market is segmented into:

HospitalsAmbulatory Surgical Center (ASCS) & Specialty ClinicsOthersBy Type, market is segmented into:

Standalone PACSIntegrated PACSBy Delivery Model, market is segmented into:

Cloud/ web based modelsOn-premise modelsOthers

The research report on Opthalmology Pacs Market provides a comprehensive analysis of the market status and development trend, including types, applications, growth, opportunities, rising technology, competitive landscape and product offerings of key players. Opthalmology Pacs Market report covers the present and past market scenarios, market development patterns, and is likely to proceed with a continuing development over the forecast period. Opthalmology Pacs Market report provides in-depth statistics and analysis available on the market status of the Opthalmology Pacs key players and is a valuable method of obtaining guidance and direction for companies and business enterprise insider considering the Opthalmology Pacs market. It contains the analysis of drivers, challenges, and restraints impacting the industry.

Global Opthalmology Pacs Market 2020 report offers a complete evaluation of the industry and contains forthcoming trend, ongoing Growth Factors, opinions, facts, past data, and static and industry data. Worldwide Opthalmology Pacs Market 2020-2026, has been structured based on an overall market analysis with information from Opthalmology Pacs industry expert. The report provides the Opthalmology Pacs market landscape and its growth prospect over the coming years. The Opthalmology Pacs report also includes a data of the key players performing in this market. The Opthalmology Pacs research study offers assessment for Global Forecast between 2020- 2026. The worldwide Opthalmology Pacs industry report executes a thorough study of the chronicled information, current and additionally upcoming business trends of Opthalmology Pacs market and future believable outcomes.

Browse the complete report @ https://www.adroitmarketresearch.com/industry-reports/opthalmology-pacs-market

Regional and Country- level Analysis Opthalmology Pacs market of different geographical areas are studied deeply and an economical scenario has been offered to support new entrants, leading market players, and investors to regulate emerging economies. The top producers and consumers focus on production, product capacity, value, consumption, growth opportunity, and market share in these key regions, covering With tables and figures helping analyze worldwide Global Opthalmology Pacs Market Forecast provides key statistics on the state of the industry and is a valuable source of guidance and direction for companies and individuals interested in the market.

Some of the key questions answered in this report:What will the market growth rate, growth momentum or acceleration market carries during the forecast period?Which are the key factors driving the Opthalmology Pacs market?What was the size of the emerging Opthalmology Pacs market by value in 2018?What will be the size of the emerging Opthalmology Pacs market in 2026?Which region is expected to hold the highest market share in the Opthalmology Pacs market?What trends, challenges and barriers will impact the development and sizing of the Global Opthalmology Pacs market?What are sales volume, revenue, and price analysis of top manufacturers of Opthalmology Pacs market?What are the Opthalmology Pacs market opportunities and threats faced by the vendors in the global Opthalmology Pacs Industry?

Contact Us :

Ryan JohnsonAccount Manager Global3131 McKinney Ave Ste 600, Dallas,TX 75204, U.S.APhone No.: USA: +1 972-362 -8199 / +91 9665341414

See the article here:
Opthalmology Pacs Market Global Innovations, Competitive Analysis, New Business Developments and Top Companies Global Forecast to 2026 - Cheshire...

Read More...

Healthcare IT Integration Market Size 2026 Global Industry Sales, Revenue, Price trends and more : Allscripts Healthcare Solutions, Inc – The Courier

December 4th, 2020 12:15 am

A new report added by Big Market Research claims that the globalHealthcare IT Integration marketgrowth is set to reach newer heights during the forecast period,2020-2026.

This report is meticulous research formulated by market professionals by deeply analyzing key driving and restraining factors, major regional market situations, major players, and size & scope of the market. The report also offers value chain analysis, Porters Five Forces model, and PEST analysis. Furthermore, the competitive landscape in different regions is elucidated in the report to assist top market players, new entrants, and investors determine investment opportunities.

Request for a sample copy of this report @https://www.bigmarketresearch.com/request-sample/3848351?utm_source=SHASHI&utm_medium=MC

The report also explains the factors boosting the market growth. The major drivers of the Healthcare IT Integration market are:

Allscripts Healthcare Solutions, Inc., Cerner Corporation, Nextgen Healthcare, Inc., Interfaceware, Inc., Epic Systems Corporation, Intersystems Corporation, Orion Health Group Limited, GE Healthcare, IBM, Corepoint Health,

In addition, the research report provides a comprehensive analysis of the key segments of the Healthcare IT Integration market. An outline of each market segment such as type, application, and region are also provided in the report.

On the basis of region, the market is evaluated across:

North America (U.S., Canada, Mexico, etc.)

Asia-Pacific (China, Japan, India, Korea, Australia, Indonesia, Taiwan, Thailand, etc.)

Europe (Germany, UK, France, Italy, Russia, Spain, etc.)

Middle East & Africa (Turkey, Saudi Arabia, Iran, Egypt, Nigeria, UAE, Israel, South Africa, etc.)

South America (Brazil, Argentina, Colombia, Chile, Venezuela, Peru, etc.)

The report outlines company profiles, product specifications and capacity, production value, and 2020-2026 market shares of key players active in the market. Moreover, the research presents the performance and recent developments of the foremost players functioning in the market. The insights delivered in the report are valuable for individuals or companies interested in investing in the Healthcare IT Integration industry. These insights might help market players in devising effective business strategies and taking the required steps to obtain a leadership position in the industry.

Shareholders, stakeholders, product managers, marketing officials, investors, and other professionals in search of detailed data on supply, demand, and future predictions of the Healthcare IT Integration market would find the report beneficial. Furthermore, we endeavor to deliver a customized report to fulfill the special requirements of our clients, on-demand.

Research Objective

To analyze and forecast the market size of global Healthcare IT Integration market.

To classify and forecast global Healthcare IT Integration market based on product, sources, application.

To identify drivers and challenges for global Healthcare IT Integration market.

To examine competitive developments such as mergers & acquisitions, agreements, collaborations and partnerships, etc., in global Healthcare IT Integration market.

To conduct pricing analysis for global Healthcare IT Integration market.

To identify and analyze the profile of leading players operating in global Healthcare IT Integration market.

The report is useful in providing answers to several critical questions that are important for the industry stakeholders such as manufacturers and partners, end users, etc., besides allowing them in strategizing investments and capitalizing on market opportunities. Key target audience are:

Manufacturers of Healthcare IT Integration

Raw material suppliers

Market research and consulting firms

Government bodies such as regulating authorities and policy makers

Organizations, forums and alliances related to Healthcare IT Integration

Highlights following key factors:

:- Business description A detailed description of the companys operations and business divisions.:- Corporate strategy Analysts summarization of the companys business strategy.:- SWOT Analysis A detailed analysis of the companys strengths, weakness, opportunities and threats.:- Company history Progression of key events associated with the company.:- Major products and services A list of major products, services and brands of the company.:- Key competitors A list of key competitors to the company.:- Important locations and subsidiaries A list and contact details of key locations and subsidiaries of the company.:- Detailed financial ratios for the past five years The latest financial ratios derived from the annual financial statements published by the company with 5 years history.

Avail the Discount on this Report @https://www.bigmarketresearch.com/request-for-discount/3848351?utm_source=SHASHI&utm_medium=MC

Table of Contents

Chapter 1. Executive Summary

Chapter 2. Global Healthcare IT Integration Market Definition and Scope

Chapter 3. Global Healthcare IT Integration Market Dynamics

Chapter 4. Global Healthcare IT Integration Market: Industry Analysis

Chapter 5. Global Healthcare IT Integration Market, by Method

Chapter 6. Global Healthcare IT Integration Market, by Style

Chapter 7. Global Healthcare IT Integration Market, by Application

Chapter 8. Global Healthcare IT Integration Market, Regional Analysis

Chapter 9. Competitive Intelligence

Chapter 10. Research Process

Tending Report :

Global Specialty PACS Market Size study, by Type (Radiology PACS, Cardiology PACS, Pathology PACS, Opthalmology PACS, Womens Health PACS, Others), by Deployment Model (On-Premises, Web/Cloud-Based), by Component (Software, Services, Hardware), by EJLN-User (Hospitals, Ambulatory Surgical Centers & Clinics, Diagnostic Imaging Centers, Others) aJLN Regional Forecasts 2020-2026

https://www.bigmarketresearch.com/report/3848352/global-specialty-pacs-market?utm_source=SHASHI&utm_medium=RR

About Us:

Big Market Research has a range of research reports from various publishers across the world. Our database of reports of various market categories and sub-categories would help to find the exact report you may be looking for.

We are instrumental in providing quantitative and qualitative insights on your area of interest by bringing reports from various publishers in one place to save your time and money. A lot of organizations across the world are gaining profits and great benefits from information gained through reports sourced by us.

Contact Us:

Mr. Abhishek Paliwal

5933 NE Win Sivers Drive, #205, Portland,

OR 97220 United States

Call answer Direct:+1-971-202-1575

Call answer Toll-Free:+1-800-910-6452

Emailhelp@bigmarketresearch.com

Read more here:
Healthcare IT Integration Market Size 2026 Global Industry Sales, Revenue, Price trends and more : Allscripts Healthcare Solutions, Inc - The Courier

Read More...

Dental Imaging Market Detailed Analysis Of Current Industry Figures With Forecasts Growth By 2026 – Cheshire Media

December 4th, 2020 12:15 am

Dental Imaging Market Forecast 2020-2026

The Global Dental Imaging Market research report provides and in-depth analysis on industry- and economy-wide database for business management that could potentially offer development and profitability for players in this market. This is a latest report, covering the current COVID-19 impact on the market. The pandemic of Coronavirus (COVID-19) has affected every aspect of life globally. This has brought along several changes in market conditions. The rapidly changing market scenario and initial and future assessment of the impact is covered in the report. It offers critical information pertaining to the current and future growth of the market. It focuses on technologies, volume, and materials in, and in-depth analysis of the market. The study has a section dedicated for profiling key companies in the market along with the market shares they hold.

The report consists of trends that are anticipated to impact the growth of the Dental Imaging Market during the forecast period between 2020 and 2026. Evaluation of these trends is included in the report, along with their product innovations.

Get a PDF Copy of the Sample Report for free @ https://dataintelo.com/request-sample/?reportId=84781

The Report Covers the Following Companies:Danaher CorporationCarestream Health, Inc. (Subsidiary Of Onex Corporation)Planmeca OySirona Dental Systems, Inc.Dentsply International, Inc.Flow Dental CorporationLed Medical Diagnostic, Inc.Midmark CorporationThe Yoshida Dental Mfg. Co., Ltd.Vatech Co. Ltd.

By Types:Extraoral ImagingIntraoral Imaging

By Applications:Dental ClinicsDental LaboratoriesDental Academic and Research InstitutesForensic Laboratories

Furthermore, the report includes growth rate of the global market, consumption tables, facts, figures, and statistics of key segments.

By Regions:

Grab Your Report at an Impressive Discount! Please visit https://dataintelo.com/ask-for-discount/?reportId=84781

Years Considered to Estimate the Market Size:History Year: 2015-2019Base Year: 2019Estimated Year: 2020Forecast Year: 2020-2026

Important Facts about Dental Imaging Market Report:

What Our Report Offers:

Make an Inquiry of This Report @ https://dataintelo.com/enquiry-before-buying/?reportId=84781

About DataIntelo:DATAINTELO has set its benchmark in the market research industry by providing syndicated and customized research report to the clients. The database of the company is updated on a daily basis to prompt the clients with the latest trends and in-depth analysis of the industry. Our pool of database contains various industry verticals that include: IT & Telecom, Food Beverage, Automotive, Healthcare, Chemicals and Energy, Consumer foods, Food and beverages, and many more. Each and every report goes through the proper research methodology, validated from the professionals and analysts to ensure the eminent quality reports.

Contact Info:

Name: Alex Mathews

Address: 500 East E Street, Ontario,

CA 91764, United States.

Phone No: USA: +1 909 545 6473

Email:[emailprotected]

Website:https://dataintelo.com

View post:
Dental Imaging Market Detailed Analysis Of Current Industry Figures With Forecasts Growth By 2026 - Cheshire Media

Read More...

New Drug Could Improve Effectiveness of Stem Cell Therapy – Pain News Network

December 4th, 2020 12:13 am

By Pat Anson, PNN Editor

Scientists have developed an experimental drug that can lure stem cells to damaged tissues and help them heal -- a discovery being touted as a major advancement in the field of regenerative medicine.

The findings, recently published in the Proceedings of the National Academy of Sciences (PNAS), could improve the effectiveness of stem cell therapy in treating spinal cord injuries, stroke, amyotrophic lateral sclerosis(ALS), Parkinsons disease and other neurodegenerative disorders. It could also expand the use of stem cells to treat conditions such as heart disease and arthritis.

The ability to instruct a stem cell where to go in the body or to a particular region of a given organ is the Holy Grail for regenerative medicine, said lead authorEvan Snyder, MD, director of theCenter for Stem Cells & Regenerative Medicineat Sanford Burnham Prebys Medical Discovery Institute in La Jolla, CA. Now, for the first time ever, we can direct a stem cell to a desired location and focus its therapeutic impact.

Over a decade ago, Snyder and his colleagues discovered that stem cells are drawn to inflammation -- a biological fire alarm that signals tissue damage has occurred. However, using inflammation as a therapeutic lure for stem cells wasnt advisable because they could further inflame diseased or damaged organs, joints and other tissue.

To get around that problem, scientists modified CXCL12 -- an inflammatory molecule that Snyders team discovered could guide stem cells to sites in need of repair to create a drug called SDV1a. The new drug works by enhancing stem cell binding, while minimizing inflammatory signals.

Since inflammation can be dangerous, we modified CXCL12 by stripping away the risky bit and maximizing the good bit, Snyder explained. Now we have a drug that draws stem cells to a region of pathology, but without creating or worsening unwanted inflammation.

To demonstrate its effectiveness, Snyders team injected SDV1a and human neural stem cells into the brains of mice with a neurodegenerative disease called Sandhoff disease. The experiment showed that the drug helped stem cells migrate and perform healing functions, which included extending lifespan, delaying symptom onset, and preserving motor function for much longer than mice that didnt receive the drug. Importantly, the stem cells also did not worsen the inflammation.

Researchers are now testing SDV1as ability to improve stem cell therapy in a mouse model of ALS, also known as Lou Gehrigs disease, which is caused by a progressive loss of motor neurons in the brain. Previous studies conducted by Snyders team found that broadening the spread of neural stem cells helps more motor neurons survive so they are hopeful that SDV1a will improve the effectiveness of neuroprotective stem cells and help slow the onset and progression of ALS.

We are optimistic that this drugs mechanism of action may potentially benefit a variety of neurodegenerative disorders, as well as non-neurological conditions such as heart disease, arthritis and even brain cancer, says Snyder. Interestingly, because CXCL12 and its receptor are implicated in the cytokine storm that characterizes severe COVID-19, some of our insights into how to selectively inhibit inflammation without suppressing other normal processes may be useful in that arena as well.

Snyders research is supported by the National Institutes of Health, U.S. Department of Defense, National Tay-Sachs & Allied Disease Foundation, Childrens Neurobiological Solutions Foundation, and the California Institute for Regenerative Medicine (CIRM).

Thanks to decades of investment in stem cell science, we are making tremendous progress in our understanding of how these cells work and how they can be harnessed to help reverse injury or disease, says Maria Millan, MD, president and CEO of CIRM. This drug could help speed the development of stem cell treatments for spinal cord injury, Alzheimers, heart disease and many other conditions for which no effective treatment exists.

Continue reading here:
New Drug Could Improve Effectiveness of Stem Cell Therapy - Pain News Network

Read More...

Repairing the Brain With Stem Cells? A Conversation With Prof. Jack Price – Being Patient

December 4th, 2020 12:13 am

Groundbreaking research in stem cells has propelled scientists understanding of neurodegenerative diseases, including Parksinsons. Could stem cell therapies one day help cure Alzheimers?

Clinical trials of stem cell therapies are now underway to repair the damaged cells of people with Parkinsons disease and age-related macular degeneration. Being Patient spoke with Jack Price, professor of developmental neurobiology at Kings College London and author of the book The Future of Brain Repair, about the potential and challenges of repairing the brain with stem cell therapy.

Being Patient: What is stem cell therapy?

Prof. Jack Price: Its the transplantation of stem cells, either directly into the brain or in a way that gives them access to the brain and influence the brain, to bring about a therapeutic effect.

Being Patient: Are there stem cells in the brain?

Prof. Jack Price: For many years, neuroscientists didnt think there were stem cells in the brain. We now know there are. We know about a population [of stem cells] thats become very important in our understanding of Alzheimers disease and in mood disorders like anxiety and depression. These are stem cells that are found in a part of the brain called the hippocampus.

But by and large, the brain doesnt have stem cells, unlike skin and other tissues in the body. The blood is the classic [example]: Theres a population of stem cells in the bone marrow that regenerates blood all the time.

Being Patient: What makes stem cells so special and why are they a focus of research?

Prof. Jack Price: The definition of stem cells is a population of cells that gives rise to other types of cells. In neural stem cells, precursor cells can make adult brain cells, nerve cells, glial cells, all the different cell types that make up the brain. If you have a disease like Alzheimers or any other neurodegenerative disease, where we know the key pathology is the loss of nerve cells, your brain doesnt normally have the ability to replace those lost brain cells. The idea was [that] if you put stem cells where the loss of brain cells has taken place, maybe those stem cells would replace the lost cells.

Being Patient: What is the potential of stem cell therapy in treating neurodegenerative diseases?

Prof. Jack Price: Theres a piece of absolutely brilliant stem cell science that was done by Shinya Yamanaka in Kyoto in 2006. He showed you could effectively take any cell through a very straightforward genetic manipulation that he discovered, [and] turn them into what we call pluripotent stem cells, which are cells that can make any cell type in the body. They also have an ability that other stem cells generally dont: They can build tissue. If you grow them in a little culture dish, they can start to make little pieces of brain called organoids or cerebroids. This was a groundbreaking technology.

In Parkinsons disease, theres enormous progress and clinical trials are underway now. We know more about the pathology of Parkinsons disease [than in Alzheimers]. The pathology of Alzheimers turns out to be quite complex, and weve had, over the years, quite a few ideas about how it worked. But [turning] those into actual therapies hasnt quite worked as we expected, and we keep having to go back and rethink whats going on in Alzheimers.

The pathology of Parkinsons disease is also difficult. Its not trivial. But at the same time, one thing is clear: a lot of the pathology is associated with the loss of a particular population of nerve cells the midbrain dopaminergic cells. We can start with these pluripotent stem cells and make them make precisely the right type of dopaminergic cell that we know is lost in Parkinsons disease.

This is built on 30 [to] 40 years of research of people trying to find exactly the right cell type to work [with] in Parkinsons disease. They had some early success and fell backwards. But this technology looks much more precise than everything anybodys ever tried before.

In age-related macular degeneration, the disease of the eye where you lose your retinal photoreceptors, there are very clever strategies now where people are using these pluripotent stem cells to make a thing called a retinal pigment epithelium. It lies behind the retina, but its what supports the photoreceptors. It turns out, thats what goes wrong in age-related macular degeneration.

Being Patient: Are there any stem cell therapy approved to treat brain disorders?

Prof. Jack Price: There are no licensed stem cell therapy for any brain disorders anywhere in the world for the simple reason [that] nobody has shown one works. There are a lot of stem cell clinics in the U.S. and somewhat fewer elsewhere who are offering cell therapies that are untested. Theyll put stem cells into you for any disorder youve got. Those cell therapies do not work.

A lot of genuine companies are trying to get these cell therapies to work in clinical trials and falling flat on their face quite often, despite their best efforts. 90% of clinical trials fail, and thats clinical trials of conventional drugs by drug companies that know what theyre doing.

What do you suppose is the chance with a stem cell therapy [that] we dont really understand how it works, [that] we dont quite know how to manufacture it properly, [and that] we dont quite know what cells we really want, of working? The chance is almost zero. These companies know that, which is why theyre not going to clinical trials.

The interview has been edited for length and clarity.

Contact Nicholas Chan at nicholas@beingpatient.com

See the rest here:
Repairing the Brain With Stem Cells? A Conversation With Prof. Jack Price - Being Patient

Read More...

Stem Cell Therapy Market Research Report Forecast to 2029 (Includes Business Impact of COVID-19) – Cheshire Media

December 4th, 2020 12:13 am

Trusted Business Insights answers what are the scenarios for growth and recovery and whether there will be any lasting structural impact from the unfolding crisis for the Stem Cell Therapy market.

Trusted Business Insights presents an updated and Latest Study on Stem Cell Therapy Market 2020-2029. The report contains market predictions related to market size, revenue, production, CAGR, Consumption, gross margin, price, and other substantial factors. While emphasizing the key driving and restraining forces for this market, the report also offers a complete study of the future trends and developments of the market.The report further elaborates on the micro and macroeconomic aspects including the socio-political landscape that is anticipated to shape the demand of the Stem Cell Therapy market during the forecast period (2020-2029).It also examines the role of the leading market players involved in the industry including their corporate overview, financial summary, and SWOT analysis.

Get Sample Copy of this Report @ Stem Cell Therapy Market Research Report Forecast to 2029 (Includes Business Impact of COVID-19)

Abstract, Snapshot, Market Analysis & Market Definition: Stem Cell Therapy MarketIndustry / Sector Trends

Stem Cell Therapy Market size was valued at USD 7.8 billion in 2018 and is expected to witness 10.2% CAGR from 2019 to 2025.

U.S. Stem Cell Therapy Market Size, By Type, 2018 & 2025 (USD Million)

Rising prevalence of chronic diseases will positively impact the stem cell therapy market growth. Cardiovascular diseases, neurological disorders and other chronic conditions have resulted in high mortality over past few years. Conventional therapeutic methods and treatments are currently replaced due to lack of efficiency and efficacy. Recently developed stem cell therapies are capable of replacing defective cells to treat diseases that has reduced morbidity drastically. Therefore, people have now started relying on stem cell therapy that has long term positive effects.

Advancements in stem cell therapy in developed regions such as North America and Europe have boosted the industry growth. Since past few years, there have been several researches carried out for stem cell therapy. Currently developed stem cell therapies have shown positive outcomes in treatment of leukemia. Similarly, due to advancements in regenerative medicine, several other chronic conditions such as muscular dystrophy and cardiovascular diseases also have been cured. Aforementioned factors have surged the industry growth. However, high cost of allogenic stem cell therapy may hamper the industry growth to some extent.

Market Segmentation, Outlook & Regional Insights: Stem Cell Therapy Market

Stem Cell Therapy Market, By Type

Allogenic stem cell therapy segment held around 39% revenue share in 2018 and it is anticipated to grow substantially during the analysis timeframe. Allogenic stem cell is available as off the shelf therapy and it is easily scalable that helps in providing treatment without delay. Moreover, the procedure includes culturing donor-derived immunocompetent cells that are highly effective in treatment of several diseases. Stem cells obtained in allogenic therapy are free of contaminating tumor cells. This reduces risk for disease recurrence that will surge its demand thereby, stimulating segment growth.

Autologous stem cell therapy segment is estimated to witness 10.1% growth over the forthcoming years. People usually prefer autologous stem cell therapy as it has minimum risk of immunological rejection. However, on introduction of allogenic stem cell therapy, demand for autologous stem cell therapy has declined as it is difficult to scale up. However, there are concerns regarding risk of cross contamination during large scale manufacturing of autologous stem cell lines that will impede segmental growth to some extent.

Stem Cell Therapy Market, By Application

The neurology segment was valued at around USD 1.6 billion in 2018 and it is estimated that it will witness significant growth over the forthcoming years. Stem cells are used to replenish the disrupted neurological cells that help in quick patient recovery. Pluripotent stem cells provide a replacement for cells and tissues to treat Alzheimers, Parkinsons disease, cerebral palsy, amyotrophic lateral sclerosis, and other neurodegenerative diseases. Thus, the pivotal role of stem cells in treating the life-threatening neurological condition will escalate segment growth.

The cardiovascular segment will witness 10% growth over the analysis timeframe. Considerable segmental growth can be attributed to development in stem cell therapies that have enhanced recovery pace in patients suffering from cardiovascular diseases. Recently developed allogeneic stem cell therapies are efficient and easily available that have reduced the mortality rates in cardiovascular patients. Above mentioned factors will propel cardiovascular segment growth in near future.

Stem Cell Therapy Market, By End-users

The hospital segment held over 56% revenue share in 2018 and it is anticipated to grow significantly in near future. The rising preference for stem cell therapies offered by hospitals proves beneficial for business growth. Hospitals have affiliations with research laboratories and academic institutes that carry out research activities for developing stem cell therapies. On the introduction and approval of any novel stem therapy, hospitals implement it immediately. Associations with research and academic institutes further help hospitals to upgrade its stem cell treatment offerings that positively impact the segmental growth.

The clinics segment is expected to grow at around 10% during the forecast timeframe. Clinics specializing in providing stem cell therapies are well-equipped with advanced medical devices and superior quality reagents required for imparting stem cell therapies. However, as clinics offer specialized stem cell therapies, their treatment cost is much higher as compared to hospitals that may reduce its preference.

Stem Cell Therapy Market, By Region

North America stem cell therapy market held around 41.5% revenue share in 2018 and it is estimated to grow substantially in near future. Increasing the adoption of novel stem cell therapies will prove beneficial for regional market growth. Moreover, favorable government initiatives have a positive impact on regional market growth. For instance, the government of Canada has initiated Strategic Innovation Fund Program that invests in research activities carried out for stem cell therapies enabling development in stem cell therapy. Above mentioned factors are expected to drive the North America market growth.

Asia Pacific stem cell therapy market is anticipated to witness 10.8% growth in the near future owing to increasing awareness amongst people pertaining to the benefits of advanced stem cell therapies. Additionally, favorable initiatives undertaken by several organizations will promote industry players to come up with innovative solutions. For instance, according to Pharma Focus Asia, members of the Asia-Pacific Economic Cooperation collaborated with Life Sciences Innovation Forum to involve professionals having expertise in stem cell therapies from academia and research centers to promote developments in stem cell research. Thus, growing initiatives by organizations ensuring the availability of new stem cell therapies will foster regional market growth.

Latin America Stem Cell Therapy Market Size, By Country, 2025 (USD Million)

Key Players, Recent Developments & Sector Viewpoints: Stem Cell Therapy Market

Key industry players in the stem cell therapy market include Astellas Pharma Inc, Cellectis, Celyad, Novadip Biosciences, Gamida Cell, Capricor Therapeutics, Cellular Dynamics, CESCA Therapeutics, DiscGenics, OxStem, Mesoblast Ltd, ReNeuron Group, and Takeda Pharmaceuticals. Chief industry players implement several initiatives such as mergers and acquisitions to sustain market competition. Also, receiving approvals for stem cell therapy products from regulatory authorities fosters the companys growth. For instance, in March 2018, the European Commission approved Takedas Alofisel that is off-the-shelf stem cell therapy. Product approval will help the company to gain a competitive advantage and capture market share.

Stem Cell Therapy Industry Viewpoint

The stem cells industry can be traced back to the 1950s. In 1959 first animals were made by in-vitro fertilization by preserving the stem cells. Till 2000, research was being carried out on stem cells to study its therapeutic effect. In 2000, fund allocations were made to research on cells derived from aborted human fetuses. In the same year, scientists derived human embryonic stem cells from the inner cell mass of blastocytes. Later, in 2010, clinical trials for human embryonic stem cell-based therapy were initiated. As technology progressed, stem cell therapy for treating cancer was developed. However, due to ethical issues, the use of stem cells for curing diseases witnessed slow growth for a few years. But as the regulatory scenario changed, people started preferring stem cell therapies due to its better efficacy. Stem cell therapy is in the developing stage and has numerous growth opportunities in developing economies with a high prevalence of chronic diseases.

Key Industry Development

In September 2020, Takeda Pharmaceutical Company Limited announced the expansion of its cell therapy manufacturing capabilities with the opening of a new 24,000 square-foot R&D cell therapy manufacturing facility at its R&D headquarters in Boston, Massachusetts. The facility provides end-to-end research and development capabilities and will accelerate Takedas efforts to develop next-generation cell therapies, initially focused on oncology with the potential to expand into other therapeutic areas.

The R&D cell therapy manufacturing facility will produce cell therapies for clinical evaluation from discovery through pivotal Phase 2b trials. The current Good Manufacturing Practices (cGMP) facility is designed to meet all U.S., E.U., and Japanese regulatory requirements for cell therapy manufacturing to support Takeda clinical trials around the world.

The proximity and structure of Takedas cell therapy teams allow them to quickly apply what they learn across a diverse portfolio of next-generation cell therapies including CAR NKs, armored CAR-Ts, and gamma delta T cells. Insights gained in manufacturing and clinical development can be quickly shared across global research, manufacturing, and quality teams, a critical ability in their effort to deliver potentially transformative treatments to patients as fast as possible.

Takeda and MD Anderson are developing a potential best-in-class allogeneic cell therapy product (TAK-007), a Phase 1/2 CD19-targeted chimeric antigen receptor-directed natural killer (CAR-NK) cell therapy with the potential for off-the-shelf use being studied in patients with relapsed or refractory non-Hodgkins lymphoma (NHL) and chronic lymphocytic leukemia (CLL). Two additional Phase 1 studies of Takeda cell therapy programs were also recently initiated: 19(T2)28z1xx CAR T cells (TAK-940), a next-generation CAR-T signaling domain developed in partnership with Memorial Sloan Kettering Cancer Center (MSK) to treat relapsed/refractory B-cell cancers, and a cytokine and chemokine armored CAR-T (TAK-102) developed in partnership with Noile-Immune Biotech to treat GPC3-expressing previously treated solid tumors.

Takedas Cell Therapy Translational Engine (CTTE) connects clinical translational science, product design, development, and manufacturing through each phase of research, development, and commercialization. It provides bioengineering, chemistry, manufacturing, and control (CMC), data management, analytical, and clinical and translational capabilities in a single footprint to overcome many of the manufacturing challenges experienced in cell therapy development.

Key Insights Covered: Exhaustive Stem Cell Therapy Market

1. Market size (sales, revenue and growth rate) of Stem Cell Therapy industry.

2. Global major manufacturers operating situation (sales, revenue, growth rate and gross margin) of Stem Cell Therapy industry.

3. SWOT analysis, New Project Investment Feasibility Analysis, Upstream raw materials and manufacturing equipment & Industry chain analysis of Stem Cell Therapy industry.

4. Market size (sales, revenue) forecast by regions and countries from 2019 to 2025 of Stem Cell Therapy industry.

Research Methodology: Stem Cell Therapy Market

Looking for more? Check out our repository for all available reports on Stem Cell Therapy in related sectors.

Quick Read Table of Contents of this Report @ Stem Cell Therapy Market Research Report Forecast to 2029 (Includes Business Impact of COVID-19)

Trusted Business InsightsShelly ArnoldMedia & Marketing ExecutiveEmail Me For Any ClarificationsConnect on LinkedInClick to follow Trusted Business Insights LinkedIn for Market Data and Updates.US: +1 646 568 9797UK: +44 330 808 0580

Read the original here:
Stem Cell Therapy Market Research Report Forecast to 2029 (Includes Business Impact of COVID-19) - Cheshire Media

Read More...

How Stem Cell Therapy Market Will Dominate In Coming Years? Report Covering Products, Financial Information, Developments, Swot Analysis And…

December 4th, 2020 12:13 am

The Global Stem Cell Therapy Market analysis report published on IndustryGrowthInsights.com is a detailed study of market size, share and dynamics covered in XX pages and is an illustrative sample demonstrating market trends. This is a latest report, covering the current COVID-19 impact on the market. The pandemic of Coronavirus (COVID-19) has affected every aspect of life globally. This has brought along several changes in market conditions. The rapidly changing market scenario and initial and future assessment of the impact is covered in the report. It covers the entire market with an in-depth study on revenue growth and profitability. The report also delivers on key players along with strategic standpoint pertaining to price and promotion.

Get FREE Exclusive PDF Sample Copy of This Report: https://industrygrowthinsights.com/request-sample/?reportId=168110

The Global Stem Cell Therapy Market report entails a comprehensive database on future market estimation based on historical data analysis. It enables the clients with quantified data for current market perusal. It is a professional and a detailed report focusing on primary and secondary drivers, market share, leading segments and regional analysis. Listed out are key players, major collaborations, merger & acquisitions along with upcoming and trending innovation. Business policies are reviewed from the techno-commercial perspective demonstrating better results. The report contains granular information & analysis pertaining to the Global Stem Cell Therapy Market size, share, growth, trends, segment and forecasts from 2020-2026.

With an all-round approach for data accumulation, the market scenarios comprise major players, cost and pricing operating in the specific geography/ies. Statistical surveying used are SWOT analysis, PESTLE analysis, predictive analysis, and real-time analytics. Graphs are clearly used to support the data format for clear understanding of facts and figures.

Customize Report and Inquiry for The Stem Cell Therapy Market Report: https://industrygrowthinsights.com/enquiry-before-buying/?reportId=168110

Get in touch with our sales team, who will guarantee you to get a report that suits your necessities.

Primary research, interviews, news sources and information booths have made the report precise having valuable data. Secondary research techniques add more in clear and concise understanding with regards to placing of data in the report.

The report segments the Global Stem Cell Therapy Market as:Global Stem Cell Therapy Market Size & Share, by Regions

Global Stem Cell Therapy Market Size & Share, by ProductsAutologousAllogeneicStem Cell Therap

Global Stem Cell Therapy Market Size & Share, ApplicationsMusculoskeletal DisorderWounds & InjuriesCorneaCardiovascular DiseasesOthers

Key PlayersOsiris TherapeuticsNuVasiveChiesi PharmaceuticalsJCR PharmaceuticalPharmicellMedi-postAnterogenMolmedTakeda (TiGenix)Stem Cell Therap

Avail the Discount on this Report @ https://industrygrowthinsights.com/ask-for-discount/?reportId=168110

IndustryGrowthInsights offers attractive discounts on customization of reports as per your need. This report can be personalized to meet your requirements. Get in touch with our sales team, who will guarantee you to get a report that suits your necessities.

About IndustryGrowthInsights:INDUSTRYGROWTHINSIGHTS has set its benchmark in the market research industry by providing syndicated and customized research report to the clients. The database of the company is updated on a daily basis to prompt the clients with the latest trends and in-depth analysis of the industry. Our pool of database contains various industry verticals that include: IT & Telecom, Food Beverage, Automotive, Healthcare, Chemicals and Energy, Consumer foods, Food and beverages, and many more. Each and every report goes through the proper research methodology, validated from the professionals and analysts to ensure the eminent quality reports.

Contact Info: Name: Alex MathewsAddress: 500 East E Street, Ontario, CA 91764, United States.Phone No: USA: +1 909 545 6473Email: [emailprotected]Website: https://IndustryGrowthInsights.com

See original here:
How Stem Cell Therapy Market Will Dominate In Coming Years? Report Covering Products, Financial Information, Developments, Swot Analysis And...

Read More...

Stem Cell Therapy Market to Witness Robust Expansion Throughout the Forecast Period 2020 2025 – The Haitian-Caribbean News Network

December 4th, 2020 12:13 am

Global Stem Cell Therapy Market 2020-2025 Introduction and Scope:

This new advanced research study and presentation on the global Stem Cell Therapy Market is ready to provide you with amazing market-related details that have a significant impact on your growth. In this report, readers will find a variety of information about regional developments, including manufacturer activity, technological leaps, new government policies affecting industry operations, and country-specific growth milestones indicating a healthy growth trajectory for the global Stem Cell Therapy Market. The report also includes a variety of data that manages new M&A proposals, commercial ventures and operations, facility expansion coverage, geographic diversification, etc. that players consider to set short and long term business goals across the growth curve. Additional details regarding the competitive environment, industry pioneers, emerging companies, versatile market participants and investors were religiously emphasized.

In addition, the report has an optimal reference to the revenue-generating potential of each sector and has been evaluated to encourage the right investment. The details of geographic regions and the likelihood of favorable consumer response in each region are closely tracked to identify the growth hotspots of the global Stem Cell Therapy Market during the forecast period 2020-25.

Sample PDF Brochure with Covid-19 Updates @ https://www.adroitmarketresearch.com/contacts/request-sample/691?utm_source=re

The report also provides ample information about the competitive landscape with vendor positioning and various details on the activities that remain essential growth catalysts. In addition to providing a detailed overview of the current market scenario, this section of the report also includes various details on the overall ecosystem, key trends, market catalysts, threats and challenges that significantly affect the monetization of the Stem Cell Therapy Market.

After continuing observations and research initiatives, this new research presentation on the global Stem Cell Therapy Market was recently released in order to optimally clean this global market to draw important conclusions.

The various research approaches included in this Stem Cell Therapy Market report compilation provide an internal report on the market size and growth trends, showing new trends and developments across multiple geographic tiers such as global and regional hubs as well as regional growth pockets.

Browse the complete report Along with TOC @ https://www.adroitmarketresearch.com/industry-reports/stem-cell-therapy-market?utm_source=re

Frequently Asked Questions: Global Stem Cell Therapy Market

1. In terms of product and application based segmentation, which segment is likely to remain most promising?2. Based on concurrent developments, which trends are likely to remain most dominant through the forecast span?3. Considering the pandemic crisis and other associated alterations, what could be the most relevant market projections?4. Who would continue to remain atop the growth curve in global Stem Cell Therapy Market through the forecast years?5. What are the top threats and challenges identified in the Stem Cell Therapy Market?

Stem Cell Therapy Market Segmentation

Type Analysis of Stem Cell Therapy Market:

Based on cell source, the market has been segmented into,

Adipose Tissue-Derived Mesenchymal SCsBone Marrow-Derived Mesenchymal SCsEmbryonic SCsOther Sources

Applications Analysis of Stem Cell Therapy Market:

Based on therapeutic application, the market has been segmented into,

Musculoskeletal DisordersWounds & InjuriesCardiovascular DiseasesGastrointestinal DiseasesImmune System DiseasesOther Applications

Reasons to read this report:

1. It helps you understand the key product segments and their future.2. It provides accurate analysis of changing competitive dynamics and allows you to stay ahead of your competitors.3. It helps you make informed business decisions by having complete insight into the market and performing in-depth analysis of market segments.

Make An Enquiry About This Report @ https://www.adroitmarketresearch.com/contacts/enquiry-before-buying/691?utm_source=re

About Us :

Adroit Market Research is an India-based business analytics and consulting company incorporated in 2018. Our target audience is a wide range of corporations, manufacturing companies, product/technology development institutions and industry associations that require understanding of a markets size, key trends, participants and future outlook of an industry. We intend to become our clients knowledge partner and provide them with valuable market insights to help create opportunities that increase their revenues. We follow a code Explore, Learn and Transform. At our core, we are curious people who love to identify and understand industry patterns, create an insightful study around our findings and churn out money-making roadmaps.

Contact Us :

Ryan JohnsonAccount Manager Global3131 McKinney Ave Ste 600, Dallas,TX75204, U.S.A.Phone No.: USA: +1 972-362 -8199/ +91 9665341414

Read the original post:
Stem Cell Therapy Market to Witness Robust Expansion Throughout the Forecast Period 2020 2025 - The Haitian-Caribbean News Network

Read More...

Stem Cell Therapy Market Expansion Projected to Gain an Uptick During 2020-2027 – The Haitian-Caribbean News Network

December 4th, 2020 12:13 am

Verified Market Research have recently published a new report on the global Stem Cell Therapy market. The study provides profound insights into updated market events and market trends. This, in turn, helps one in better comprehending the market factors, and strongly they influence the market. Also, the sections related to regions, players, dynamics, and strategies are segmented and sub-segmented to simplify the actual conditions of the industry.

The study is updated with the impacts of the coronavirus and the future analysis of the industrys trends. This is done to ensure that the resultant predictions are most accurate and genuinely calculated. The pandemic has affected all industries, and this report evaluates its impact on the global market.

Global Stem Cell Therapy Market was valued at USD 117.66 million in 2019 and is projected to reach USD 255.37 million by 2027, growing at a CAGR of 10.97% from 2020 to 2027.

The report also emphasizes the initiatives undertaken by the companies operating in the market including product innovation, product launches, and technological development to help their organization offer more effective products in the market. It also studies notable business events, including corporate deals, mergers and acquisitions, joint ventures, partnerships, product launches, and brand promotions.

Leading Stem Cell Therapy manufacturers/companies operating at both regional and global levels:

The report also inspects the financial standing of the leading companies, which includes gross profit, revenue generation, sales volume, sales revenue, manufacturing cost, individual growth rate, and other financial ratios.

Dominant participants of the market analyzed based on:

The competitors are segmented into the size of their individual enterprise, buyers, products, raw material usage, consumer base, etc. Additionally, the raw material chain and the supply chain are described to make the user aware of the prevailing costs in the market. Lastly, their strategies and approaches are elucidated for better comprehension. In short, the market research report classifies the competitive spectrum of this global Stem Cell Therapy industry in elaborate detail.

Key highlights of the report:

Market revenue splits by most promising business segments by type, by application, and any other business segment if applicable within the scope of the global Stem Cell Therapy market report. The country break-up will help you determine trends and opportunities. The prominent players are examined, and their strategies analyzed.

1.Stem Cell Therapy Market, By Cell Source:

Adipose Tissue-Derived Mesenchymal Stem Cells Bone Marrow-Derived Mesenchymal Stem Cells Cord Blood/Embryonic Stem Cells Other Cell Sources

2.Stem Cell Therapy Market, By Therapeutic Application:

Musculoskeletal Disorders Wounds and Injuries Cardiovascular Diseases Surgeries Gastrointestinal Diseases Other Applications

3.Stem Cell Therapy Market, By Type:

Allogeneic Stem Cell Therapy Market, By Application Musculoskeletal Disorders Wounds and Injuries Surgeries Acute Graft-Versus-Host Disease (AGVHD) Other Applications Autologous Stem Cell Therapy Market, By Application Cardiovascular Diseases Wounds and Injuries Gastrointestinal Diseases Other Applications

This Stem Cell Therapy report umbrellas vital elements such as market trends, share, size, and aspects that facilitate the growth of the companies operating in the market to help readers implement profitable strategies to boost the growth of their business. This report also analyses the expansion, market size, key segments, market share, application, key drivers, and restraints.

Insights into the Stem Cell Therapy market scenario:

Moreover, the report studies the competitive landscape that this industry offers to new entrants. Therefore, it gives a supreme edge to the user over the other competitors in the form of reliable speculations of the market. The key developments in the industry are shown with respect to the current scenario and the approaching advancements. The market report consists of prime information, which could be an efficient read such as investment return analysis, trends analysis, investment feasibility analysis and recommendations for growth.

The data in this report presented is thorough, reliable, and the result of extensive research, both primary and secondary. Moreover, the global Stem Cell Therapy market report presents the production, and import and export forecast by type, application, and region from 2020 to 2027.

Customization of the Report:

Verified Market Research also provides customization options to tailor the reports as per client requirements. This report can be personalized to cater to your research needs. Feel free to get in touch with our sales team, who will ensure that you get a report as per your needs.

Thank you for reading this article. You can also get chapter-wise sections or region-wise report coverage for North America, Europe, Asia Pacific, Latin America, and Middle East & Africa.

To summarize, the global Stem Cell Therapy market report studies the contemporary market to forecast the growth prospects, challenges, opportunities, risks, threats, and the trends observed in the market that can either propel or curtail the growth rate of the industry. The market factors impacting the global sector also include provincial trade policies, international trade disputes, entry barriers, and other regulatory restrictions.

Verified Market Dashboard:

VMI is BI powered Database that helps thousands of companies globally gather insights on over 20,000+ emerging and niche markets helping them make critical revenue impacting decisions. VMI assists your organization in planning for the future along with providing a holistic competitive landscape with overall market potential and in depth market share analysis broken down by Region, Country, Segments and key market leaders. VMRs database leverages its years of data gathering ability providing insights on trends and helping you with accurate future forecasts for your market research needs.

About us:

Verified Market Research is a leading Global Research and Consulting firm servicing over 5000+ customers. Verified Market Research provides advanced analytical research solutions while offering information enriched research studies. We offer insight into strategic and growth analyses, Data necessary to achieve corporate goals, and critical revenue decisions.

Our 250 Analysts and SMEs offer a high level of expertise in data collection and governance use industrial techniques to collect and analyze data on more than 15,000 high impact and niche markets. Our analysts are trained to combine modern data collection techniques, superior research methodology, expertise, and years of collective experience to produce informative and accurate research.

Contact us:

Mr. Edwyne Fernandes

US: +1 (650)-781-4080UK: +44 (203)-411-9686APAC: +91 (902)-863-5784US Toll-Free: +1 (800)-7821768

Email: [emailprotected]

Read the original post:
Stem Cell Therapy Market Expansion Projected to Gain an Uptick During 2020-2027 - The Haitian-Caribbean News Network

Read More...

Global Stem Cell Therapy Market Detailed Analysis Of Current Industry Figures With Forecasts Growth By 2027 – The Haitian-Caribbean News Network

December 4th, 2020 12:12 am

Coherent Market Insights Presents GlobalStem Cell Therapy MarketSize, Status and Forecast 2020-2027 New Document to its Studies Database

This report, which has been published, is having a meaningful Stem Cell Therapy market insight. It casts some lights on industry products and services. Along with those product applications, it also examined whether it reaches up to the end-users or not. This report on this Stem Cell Therapy market has given an overall view of the recent technologies used and technological improvements. It also focuses on recent industry trends and which products are quite demanding from a customers perspective. This report is focused on every aspect of the forecast year 2027.

This report is representing a whole market scenario on a global basis. In this report, we can also find the analysis growth of industries. Through this report, we can easily interpreter the level of market competition, different pricing models, the latest market trends, customer demand, etc. This report acknowledges the revenue model and market expansion of this Stem Cell Therapy market. If you want to get that full market information, then this report can help you. It also gives a comprehensive knowledge about the demand and supply graph. Suppose that demand curves moved downward, then from this report, you can know about those factors responsible for its decline.

Get 15% Free Customization on this Report: https://www.coherentmarketinsights.com/insight/request-customization/2848

Competitive Landscape and Stem Cell Therapy Market Share Analysis

Stem Cell Therapy market competitive landscape provides details and data information by players. The report offers comprehensive analysis and accurate statistics on revenue by the player for the period 2016-2020. It also offers detailed analysis supported by reliable statistics on revenue (global and regional level) by players for the period 2016-2020. Details included are company description, major business, company total revenue and the sales, revenue generated in Stem Cell Therapy business, the date to enter into the Stem Cell Therapy market, Stem Cell Therapy product introduction, recent developments, etc.

Some of the key players/Manufacturers involved in the Stem Cell Therapy MarketMagellan, Medipost Co., Ltd, Osiris Therapeutics, Inc., Kolon TissueGene, Inc., JCR Pharmaceuticals Co., Ltd., Anterogen Co. Ltd., Pharmicell Co., Inc., and Stemedica Cell Technologies, Inc.

Research and Methodology

For the research, the Stem Cell Therapy markets research teams are adopted various high-end techniques. Industry best analysts are worked on this report. They collected data from various reliable sources and have taken samples of different market segments. They utilize both qualitative and quantitative data in this report. All data are based on primary sources, which are focused on the assessment year 2020-2027. For wise decision-making, they have also done SWOT analysis, which can also help them know their predicted future results. This report also helps to develop Stem Cell Therapy market growth by improvising its strategic models.

Detailed Segmentation:

By Cell Source:

By Application:

If opting for the Global version of Stem Cell Therapy Market analysis is provided for major regions as follows:

North America (The US, Canada, and Mexico)

Europe (the UK, Germany, France, and Rest of Europe)

Asia Pacific (China, India, and Rest of Asia Pacific)

Latin America (Brazil and Rest of Latin America)

Middle East & Africa (Saudi Arabia, the UAE, South Africa, and Rest of Middle East & Africa)

Key Benefits:

This study gives a detailed analysis of drivers and factors limiting the market expansion of Stem Cell Therapy

The micro-level analysis is conducted based on its product types, end-user applications, and geographies

Porters five forces model gives an in-depth analysis of buyers and suppliers, threats of new entrants & substitutes and competition amongst the key market players

By understanding the value chain analysis, the stakeholders can get a clear and detailed picture of this Stem Cell Therapy market

Buy This Complete A Business Report:https://www.coherentmarketinsights.com/insight/buy-now/2848

Reasons to Buy a Full Report In depth analysis by industry experts Use of data triangulation method for examining the various aspects of the market Detailed profiling of the major competitors in the market A complete overview of the market landscape Computed Annual Growth Rate is calculated for period, 2020-2027

Table of Contents

Report Overview: It includes the Stem Cell Therapy market study scope, players covered, key market segments, market analysis by application, market analysis by type, and other chapters that give an overview of the research study.

Executive Summary: This section of the report gives information about Stem Cell Therapy market trends and shares, market size analysis by region and analysis of global market size. Under market size analysis by region, analysis of market share and growth rate by region is provided.

Profiles of International Players: Here, key players of the Stem Cell Therapy market are studied on the basis of gross margin, price, revenue, corporate sales, and production. This section gives a business overview of the players and shares their important company details.

Regional Study: All of the regions and countries analyzed in the Stem Cell Therapy market report is studied on the basis of market size by application, the market size by product, key players, and market forecast.

Actual Numbers & In-Depth Analysis, Business opportunities, Market Size Estimation Available in Full Report.

Contacts US:

Mr. ShahCoherent Market Insights,1001 4th Ave,#3200 Seattle, WA 98154, U.S.Phone: US +1-206-701-6702/UK +44-020 8133 4027Email:[emailprotected]

More:
Global Stem Cell Therapy Market Detailed Analysis Of Current Industry Figures With Forecasts Growth By 2027 - The Haitian-Caribbean News Network

Read More...

Page 274«..1020..273274275276..280290..»


2025 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick