header logo image


Page 7«..6789..»

Archive for the ‘Stem Cell kidney Failure’ Category

Stem Cell Implant Is Being Trialled To Cure" Type 1 Diabetes – IFLScience

Thursday, August 10th, 2017

A groundbreaking attempt to"cure" Type 1 diabetes with stem cells began last week. Embryonic stem cell implants were given to two people, one in the US and one in Canada, with high-risk Type 1 diabetes. The researchers hope that this willhelp the patients manage the condition.

The stem cells, developed by private company ViaCyte, are implanted underneath the patient's forearm, where they take about three months to mature into islet cells. In the pancreas, these cells are responsible for the production of insulin. In people with Type 1 diabetes, these cells are attacked by the bodys own immune system.

If it works, we would call it a functional cure, Paul Laikind of Viacyte told New Scientist. Its not truly a cure because we wouldnt address the autoimmune cause of the disease, but we would be replacing the missing cells.

A smaller implant has already been trialled on 19 people for safety and the company expects to extend the trial to 40 more people later this year, in order to understand both the safety and efficacy of the full-size implant. ViaCyte would like to get preliminary results during the first half of 2018 and to know if the system works between six and12 months later.

Islet transplants have been used to successfully treat patients with unstable, high-risk Type 1 diabetes, but the procedure has limitations, including a very limited supply of donor organs and challenges in obtaining reliable and consistent islet preparations, trial investigator James Shapiro, from the University of Alberta, said in a statement. An effective stem cell-derived islet replacement therapy would solve these issues and has the potential to help a greater number of people.

If a success, the implant will improve the lives of the patients as they wont have to closely monitor their blood levels or inject insulin, but there is a trade-off. They will have to take immunosuppressive drugs, so that their bodies dont attack the newly implanted cells. This iswhy the procedure is targeted atpeople who are at ahigher risk.

Researchers estimate that 140,000 people in Canada and the US are currently suffering from high-risk Type 1 diabetes. The condition can lead to severe episodes of hypoglycemia in the short term and heart disease, stroke, and kidney disease (among others) in thelong term.

[H/T:New Scientist]

See the original post here:
Stem Cell Implant Is Being Trialled To Cure" Type 1 Diabetes - IFLScience

Read More...

High school student gets an early start in stem cell research at USC – USC News

Wednesday, August 2nd, 2017

Even though Richard Lopez is still in high school, he can already tell you a thing or two about the ureteric bud, the metanephric mesenchyme and the developing kidney.

More impressively, he was familiar with these terms before starting his summer internship in the lab of Andy McMahon, kidney researcher and director of the Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC.

I knew I was going to come here, Lopez said. So from December on, I was just reading papers that were written by Dr. McMahons lab. And so I read about the development of the kidney, kidney organoids, experimental methods like in situ hybridization, immunohistochemistry, all that stuff. Im really glad I did all of that because now that Im here, I understand whats going on.

Lopez undertook this intense preparation as part of the Science Research Program at his Connecticut boarding school, Choate Rosemary Hall. In addition to familiarizing him with the McMahon labs research, the program provided experience with useful molecular biology techniques, ranging from gel electrophoresis to polymerase chain reaction.

Lopez didnt start his high school career at Choate. Growing up in Lennox near the Los Angeles International Airport, he attended local public schools until his sophomore year in high school. At that point, his exceptional scores on the California Standardized Test attracted the attention of the Young Eisner Scholar program, which empowers underserved students to fulfill their potential.

As an Eisner Scholar, he earned both admission and a full scholarship to attend Choate. But the decision to leave home wasnt easy.

I was terrified at first, leaving everything behind, he said. I talked to my mom about it, and at first she was hesitant because I was born and raised here, and Im the only child. But then she realized that this is an amazing opportunity, and I cant let it go by.

Lopez recalls that Choate was initially in a huge culture shock from the occasional Maserati to the international student body to the exceptional academic opportunities such as the Science Research Program that brought him to USC.

In the McMahon lab, Lopez has learned about the molecular signals that drive the branching development of the kidney, and he has practiced a wide range of lab techniques.

Im really excited about science because I know its potential.

Richard Lopez

Im really excited and passionate about science because I know its potential, he said. If you pair that with math, you have no boundaries. If you look at the lab where Im working right now creating kidney organoids, learning about kidney development, these kinds of things can solve really burdensome illnesses that are fatal to some people, like end-stage renal disease and polycystic kidney disease.

To get to the lab every day, Lopez bike commutes a total of 32 miles from his home in Lennox to USCs Health Sciences Campus. Hes run the Los Angeles Marathon once and the San Francisco Marathon twice. In November, hes planning to travel to Florida to celebrate his 18th birthday with his first Ironman Triathlon a 2.4-mile swim, 112-mile bike ride and 26.2-mile run.

Hes participating in these events not only for fun and fitness, but also as a way to give back. Hes currently raising sponsorship money for the Partnership Scholars Program, which provides underserved junior high and high school students with educational and cultural experiences, ranging from theatergoing to restaurant outings to college tours. His goal is to raise $54,000 to fund three new scholars.

I was very lucky, he said. So I want to raise money for the scholarships that have helped me out along the way.

More stories about: Research, Stem Cells

Here is the original post:
High school student gets an early start in stem cell research at USC - USC News

Read More...

UW-Madison scientists grow functional artery cells from stem cells – Madison.com

Wednesday, July 12th, 2017

In a step toward one of stem cell sciences chief goals, UW-Madison researchers have grown functional human artery cells that helped lab mice survive heart attacks.

The development, from the lab of stem cell pioneer James Thomson, could help scientists create arteries to use in bypass surgeries for cardiovascular disease, the nations top killer. Several challenges remain, however, and studies in people are years away.

This work provides valuable proof that we can eventually get a reliable source for functional arterial endothelial cells and make arteries that perform and behave like the real thing, Thomson said in a statement.

The research, reported Monday in the journal Proceedings of the National Academy of Sciences, is part of a federally funded effort at UW-Madison to create artery banks for cardiovascular surgery from universally compatible donors.

In a related project, other UW-Madison researchers are testing three-dimensional heart patches of heart muscle cells, grown from stem cells, in pigs. The goal is to replace diseased or damaged heart tissue in humans.

Since Thomson became the first scientist to successfully grow human embryonic stem cells in a lab in 1998, researchers around the world have been coaxing the universal cells into various cell types heart, pancreas, kidney, brain to develop therapies and better understand diseases.

Today, many researchers use cells reprogrammed to their embryonic state from mature cells known as induced pluri- potent stem, or iPS, cells as the raw material. Thomson helped discover iPS cells in 2007.

Many labs can convert embryonic stem cells or iPS cells into specific cell types, but developing specialized cell lines that are pure, functional and robust has been a challenge.

Thomson and his team set out to find a recipe for growing artery cells that would really function like arteries.

The researchers used two new techniques: single-cell RNA sequencing to identify genes highly expressed in cells that initiate artery development, and CRISPR-Cas9 gene editing to evaluate the function of the genes.

They found that five small molecules and growth factors are needed to encourage iPS cells to become functional artery cells. To their surprise, they discovered that insulin, a common growth factor that had been used before in trying to grow artery cells, actually inhibits such growth.

They used their recipe to make artery cells, and tested the cells in mice that had their left coronary arteries tied off to mimic heart attacks. Four weeks later, 83 percent of mice treated with the cells were alive, compared to 33 percent of mice that didnt get the cells.

We can use those cells to further create tissue-engineered arteries for bypass surgeries, said Jue Zhang, a scientist in Thomsons lab at the Morgridge Institute for Research and lead author of the study.

Developing off-the-shelf bypasses for surgery is the goal of an $8 million, seven-year grant UW-Madison received last year from the National Heart, Lung and Blood Institute to create universal artery banks.

The blood vessels of many cardiovascular disease patients arent suitable for use as bypasses, doctors say, and growing bypasses from individual patients stem cells would be timely and expensive. The hope is to use iPS cells from a rare population of genetically compatible donors to grow arteries anyone could use.

UW-Madison scientists, including engineers Tom Turng and Naomi Chesler and pathologist Igor Slukvin at the Wisconsin National Primate Research Center, plan to grow artery cells on scaffolds and test them in monkeys. If successful, the cells would be produced for human studies at the Waisman Biomanufacturing facility on campus.

The heart patches involve another $8.6 million, seven-year National Institutes of Health grant, shared with the University of Alabama-Birmingham and Duke University.

The patches involve three types of heart cells, derived from iPS cells, said Dr. Tim Kamp, a UW-Madison cardiologist and co-director of the universitys Stem Cell and Regenerative Medicine Center.

In studies in pigs, getting the patches to connect and survive when transplanted to pig hearts after heart attacks remains a challenge, Kamp said. Immune tolerance of the human grafts in pigs is another concern, he said.

But if such hurdles can be overcome, tests in humans could follow.

View original post here:
UW-Madison scientists grow functional artery cells from stem cells - Madison.com

Read More...

Kadmon ROCKing some positive results – BioPharma Dive

Wednesday, July 12th, 2017

Dive Brief:

After a disappointing initial public offering and a tough debt load looming for 2018 and 2019, Kadmon spent the latter half of 2016 putting its house in order, with a credit agreement renegotiation, staff cuts and optimization of its sales strategies. While these changes work in the background, in the foreground the company spent its R&D day yesterday celebrating some good news for KD025, its Rho-associated coiled-coil kinase 2 (ROCK2) inhibitor, which has shown similar efficacy to other drugs in development such as Imbruvica (ibrutinib) and Jakafi (ruxolitinib).

"These positive interim findings indicate activity and a favorable safety profile of KD025 in cGVHD, a fatal disease with no approved therapies,"said John Ryan,CMO of Kadmon. Steroids are the current standard therapy for cGVHD and have severe side effects associated with long-term use. We are pleased to see that the majority of patients in the first cohort have been able to reduce their steroid doses, indicating that KD025 potentially offers a well-tolerated treatment option for cGVHD patients."

The company will roll out further data for the other two cohorts, with more results on twice-daily dosing at 200 mg, and data on once daily dosing at 400 mg, during 2018. According to Jefferies analysts, Kadmon will seek breakthrough designation for KD025 by the end of 2017, and begin a clinical trial in newly diagnosed cGVHD in early 2018.

KD025 is also being assessed in a placebo-controlled Phase 2 clinical trial in moderate to severe psoriasis, and an open-label Phase 2 clinical trial in idiopathic pulmonary fibrosis. The company's pipelineincludes tesevatinib, in Phase 2 for non-small cell lung cancer and glioblastoma, and in Phase 2 and 3 for polycystic kidney disease.

Chronic graft-versus-host disease (cGVHD) is a complication of allogeneic bone marrow transplantation and hematopoietic stem cell transplantation, often used to treat myeloma and leukemia. Symptoms affect skin, mouth, liver, eyes, gastrointestinal tract, vagina, esophagus, musculoskeletal tissue and lungs. While the global GVHD market is still a relatively small one, worth around $360 million in 2016, rising to an estimated $640 million by 2026, according to Visiongain.cGVHD affects up to 50% of patients who survive longer than three months post-transplant and has a major impact on quality of life in otherwise successfully treated patients.

More:
Kadmon ROCKing some positive results - BioPharma Dive

Read More...

Kidney disease: how could stem cells help? | Eurostemcell

Wednesday, July 5th, 2017

The kidneys are towards the back of the body, roughly 10 cm above the hipbones and just below the ribcage. They are the bodys filtering units, maintaining a safe balance of fluid, minerals, salts and other substances in the blood. They produce urine to remove waste and harmful substances from the body. They also produce several hormones: erythropoietin (EPO), which acts on the bone marrow to increase the production of red blood cells; calcitriol (active Vitamin D3), which promotes absorption and use of calcium and phosphate for healthy bones and teeth; and the enzyme renin, which is involved in monitoring and controlling blood pressure.

The key working component of the kidney is the nephron.

The nephron - the functional unit of the kidney:The best evidence so far for stem cells in the adult kidney suggests they might be found in the blue area, called the urinary pole.

The nephron is made up of:

Microscope image of kidney tissue showing tubules. One tubule is highlighted to show epithelial cells (blue), cell nuclei (green) and the tubule lumen (dark center).

See the article here:
Kidney disease: how could stem cells help? | Eurostemcell

Read More...

UW-Madison scientists, inspired by old bones, find new strategy for … – Madison.com

Wednesday, July 5th, 2017

Protein-based drugs are increasingly used to treat bone disorders, kidney disease, wounds, arthritis and cancer. But the proteins frequently degrade, limiting their therapeutic potential and sometimes causing immune reactions.

UW-Madison scientists, inspired by proteins found intact in centuries-old human bones, created a mineral coating that mimics bone and appears to keep proteins stable.

Whats needed is a delivery system that remains localized, releases the protein over an extended time frame and keeps the protein active, said William Murphy, a UW-Madison professor of biomedical engineering.

Murphy and his colleagues, including Xiaohua Yu of the UW School of Medicine and Public Health, reported on their mineral coating constructed at the miniature level of biology known as the nanoscale in this weeks edition of the journal Advanced Materials.

In a lab dish, the mineral coating released a protein, called basic fibroblast growth factor, which remained active for more than a month. When the protein was released by a commonly used system made with polymers, or plastics and rubber, it stayed active for less than a week.

In rabbits, the scientists repaired Achilles tendon tears by stitching together the severed portions of the tendon. Sutures lined with mineral coating that released two growth factors healed the injury better than regular sutures, Murphy said.

Even after the mineral coatings were subject to harmful solvents in the lab dish and sterilization of the sutures conditions that can cause proteins to degenerate the proteins remained intact.

We really can hit these proteins with a sledgehammer, so to speak, and they remain protected by the mineral coating, Murphy said.

Murphy and his colleagues knew about archaeological discoveries of growth factors and other proteins preserved in human teeth and bones from the Middle Ages. But it was a 2010 report about DNA extracted from a 19,000-year-old emu eggshell in Australia that really gave him the idea for medical applications.

It triggered a greater interest in how powerful these calcified tissues can be for stabilizing biologic molecules, said Murphy, co-director of UW-Madisons Stem Cell and Regenerative Medicine Center. If we could recreate some critical aspects of mineralized tissues, they may serve as a template for protein stabilization.

He and his team created the coating by soaking a special surface in a solution containing mineral ions found in human blood, and growing crystals. Its not all that different from a really simple crystal growth kit that a grammar school student might buy, he said.

The scientists are now using the mineral coating in rodent studies of protein therapies for rheumatoid arthritis.

Other applications could include knee and hip implants and drugs for cancer, wounds and bone disorders. The system might also help improve drugs such as erythropoietin, or EPO, used to boost red blood cells damaged by kidney disease. The drugs unstable proteins can cause immune system reactions.

If proteins could be better stabilized in drug delivery, patients might need injections only once every month or two instead of every day or week, Murphy said.

Originally posted here:
UW-Madison scientists, inspired by old bones, find new strategy for ... - Madison.com

Read More...

Omeros: Next Big Thing In Biotech – Seeking Alpha

Wednesday, July 5th, 2017

Omeros Corporation (OMER) is a biopharmaceutical company which develops and commercialize both small-molecule and protein therapeutics for the treatment of inflammation, coagulopathies and disorders of the central nervous system. Omeros has one of the most diverse pipelines and recently emerged as a strong investment target. The company has one marketed product and four drug candidates in clinical stage with various others in preclinical stage in their pipeline. A company with 12 drug candidate under development in their clinical and pre clinical pipeline has lot of potential and could turn out to be a company with blockbuster portent in the next couple of years.

Omeros first marketed product Omidria, a 1% phenylephrine and 0.3% ketorolac injection is a part of its proprietary PharmacoSurgery platform. It is primarily designed to prevent the intraoperative miosis (pupil constriction) during cataract surgery or intraocular lens (IOL) replacement and to reduce postoperative ocular pain. The main benefit from Omidria is that it prevents damage to the ocular structures, especially the iris, during surgery as a result of smaller pupil size.

Omidria is a pupil dilator that helps eye surgeons operate on lenses with fewer complications. In 2016, Omidria generated over $41 million in total sales, and in Q1 2017, it generated $12.3 million in sales. At present, nearly 3 million cataract surgeries are performed each year in the U.S. alone and assuming that it works better than any other traditional method of maintaining pupil dilation during surgery, this drug has billion dollar potential.

The pipeline for Omeros is diverse and quite extensive. OMS721 is the lead product candidate most likely to hit the market next. Currently, OMS721 is being tested in three areas; atypical hemolytic uremic syndrome (aHUS, which is a rare life threatening disorder resulting in blood clot throughout the body), hematopoietic stem cell transplant associated thrombotic microangiopathy (HSCT-TMA, a disorder triggered by stem cell transplant surgery), and immunoglobulin A nephropathy (IGAN, a disease where IgA antibodies build up in the kidneys leading to inflammation and possible kidney failure). OMS721 for treating Atypical Hemolytic Uremic Syndrome is already in the Phase 3 stage.

OMS721 is a human monoclonal antibody targeting a protein called "MASP-2.", which when inhibited can help prevent damaging immune reactions seen in the above disorders. OMS721 recently received the breakthrough therapy designation from the FDA for treating IgA Nephropathy and other renal diseases. FDAs breakthrough therapy designation enables expedited development and review of a drug candidate for the treatment of a serious or life-threatening disease. Proteinuria (uACR) is an important marker for disease progression in patients with IgA nephropathy, and improvement in proteinuria is associated with improved clinical outcomes. The company announced positive data from its Phase 2 clinical trial of OMS721 for the treatment of serious kidney disorders, including IgA nephropathy. As reported for the three patients who completed treatment, mean improvement in uACR values was 76% and mean decrease in 24-hour urine protein levels was 66%. Concurrently, daily steroid doses for all patients were substantially reduced or completely eliminated.

Omeros also is pursuing accelerated approval for OMS721 in aHUS. The FDA has already granted fast track designation for OMS721 in patients with aHUS and orphan designation for OMS721 in patients with complement-mediated TMAs, including aHUS and HSCT-TMA. The potential of the lead pipeline candidate OMS721 of Omeros seems to be even more exciting than Omidrias. Its lead clinical-stage candidate OMS721 has shown positive data from Phase 2 clinical trials in both renal disorders and hematopoietic stem cell transplant-associated thrombotic microangiopathy (HSCT-TMA). If the results achieved in small and mid-stage clinical trials can be repeated in late-stage studies, which are currently underway, Omeros could have a second drug with $1 billion per year sales potential.

A success from the company's first drug, and encouraging clinical trial results from the lead product candidate OMS721 which is in late-stage development, has produced strong movements in the company stock in the recent past. It gained close to 150 percent this year. The company has multiple drugs in its pipeline, which means that the company needs to have a steady cash flow to ensure that the development work is carried out without any hindrance.

The company has several upcoming catalysts as it progresses with the clinical trial of its lead drug candidates . The companys lead product candidate OMS721 is in Phase 3. Any positive news on this candidate will help propel the stock further. Omeros Corporation's market cap has more than doubled this year to about $1 billion but there is plenty of potential still left. If Omidria and OMS721 continue on their success to blockbuster status, the stock could give huge incentives over the next couple of years.

Disclosure: I/we have no positions in any stocks mentioned, and no plans to initiate any positions within the next 72 hours.

I wrote this article myself, and it expresses my own opinions. I am not receiving compensation for it (other than from Seeking Alpha). I have no business relationship with any company whose stock is mentioned in this article.

Excerpt from:
Omeros: Next Big Thing In Biotech - Seeking Alpha

Read More...

Benefit to assist former Litchfield paramedic – Crow River Media

Wednesday, July 5th, 2017

Knowing hes helped many people in times of medical crises, friends and family of Jesse Wadsworth want to lift up the former Litchfield EMT/paramedic in his own time of need.

Wadsworth was diagnosed with multiple myeloma, a rare bone marrow cancer, in mid-May. A former Willmar resident who recently moved to Clara City, he has worked as an EMT/paramedic in Meeker and Stearns counties for 18 years, including 13 years in Litchfield.

Wadsworth started as an EMT in Litchfield in 2003. He became a paramedic in 2008 and served as a captain for Gold Cross Ambulance in Litchfield from 2012 to 2016.

Wadsworth and his family wife, Carrie, and sons, Ben, 19, Blake, 15, and Matthew, 11 have made the Willmar area their home for the past 18 years.

Family and friends have organized a benefit dinner and silent auction, set for July 28, at the Spicer American Legion (155 Lake Ave. N.) A suggested donation of $10 will be accepted for the meal, and drinks will be available.

After his cancer diagnosis May 17, Wadsworth immediately began intraveneous and oral chemotherapy, according to his sister, Misty Maher, who is helping organize the benefit. On May 23, he was admitted to Mayo Clinic in Rochester for two weeks due to kidney failure. Along with chemotherapy, he began plasmapharisis (similar to dialysis) and blood transfusions due to anemia.

Wadsworth continues to travel weekly to Mayo Clinic for chemotherapy treatments. In a few months, after he concludes chemotherapy treatment, he will begin to receive stem cell transplants. He will be confined to a clean room at the Mayo hospital for weeks at a time, Maher said.

Wadsworth is the familys primary financial provider, but due to the serious nature of his condition and the medical treatment it necessitates, he is unable to work at this time, Maher said.

Wadsworths wife is his primary caregiver and travels with him to each appointment, staying until his release.

Maher said funds raised at the benefit will assist with medical expenses, but, just as important, she hopes his friends, family, present and past colleagues and the communities hes served will gather around him and lift him up with support, giving and prayer to help ease a burden and feel surrounded by love during this difficult time.

Maher said donations for the benefit groceries for dinner, dcor, silent auction items and cash or gift certificates are appreciated. To volunteer or to donate, contact Maher at 320-262-1498 or Wadsworths sister-in-law, Nickie Johnson, at 304-790-4668.

Read the original post:
Benefit to assist former Litchfield paramedic - Crow River Media

Read More...

Stem Cell Heart Regeneration – Turmeric Curcumin Benefits …

Sunday, November 27th, 2016

THE STEM CELL & TURMERIC RESEARCH REPORT

Few people are aware of the more than 4400 government trials that have been in progress using adult stem cellsfor more than 87 major diseases.The documented current stem cell progressis nothing short of incredible. Thefull colorChristian Wilde Stem Cell and Turmeric Research Report has been informing families of the ongoingpromise of stem cell researchandwhat exciting breakthroughs lie aheadto help your family or friends live longer healthier lives.

The growing body of scientific evidence on the benefits of turmeric is now a regular ongoing part of the report. It is now the Christian Wilde Stem Cell and Turmeric Research Report. The information is astounding.

DISEASESCOVEREDIN ISSUE ONE (issues 1-4)

Diabetes I, breast cancer, macular degeneration, retinitis pigmentosa, stroke damage, (MS) multiple sclerosis, Parkinson's, osteo and rheumatoid arthritis. The recruiting heart failure trials. Stem cells now available for animals*

DISEASES IN ISSUE TWO

Alzheimer's disease, diabetes II, spinal injury, kidney disease, HIV, cerebral palsy, autism and more ...

DISEASES IN ISSUE THREE

Lymphomas and lukemias, brain cancer, prostate cancer, bone and ligament disease, liver disease. Preventive and protective properties of turmeric/curcumin.

DISEASES IN ISSUE FOUR

Ovarian cancer, crohn's corneal regeneration, systemic Lupus. (Building organs with stem cells forhearts, tracheas, livers, kidneys.) Enrolling centers for heart failure patients and storage of umbilical cords.

DISEASES IN ISSUE FIVE (ordering issues 5-8)

Aortic Valve, Non Surgical Replacement, Stem cell trials for MS, Turmeric for pancreatic cancer, Updates on turmeric and diseases, EMBRYONIC VS ADULT STEM CELLS, Stem cells for Parkinson's AND MORE.

DISEASES IN ISSUE SIX

Fibromyalgia, Stem Cells for MD, Stem cells for arthritic damage. First success for Pulmonary hypertension. Reversing Type I and II diabetes, + Surprise interviews with trial leaders and an interview with one of17 patients actuallyhealed for(3 full years) of Relapsing, Remitting Multiple Sclerosis. Five leading cancers in US compared to India where turmeric is consumed in the diet. Arthritic damage and stem cells. Dr. Paul Ridker from Harvard (father of inflammation) makes Time Magazine's top 10.

DISEASES INISSUE 7

A comprehensive study and report on howall types of cancer are benefiting from turmeric/curcumin both as a preventive and for existing cancer. In order to bring you the most accurate scientific information on this spice and how it does what it does, the world leader in the research who has conducted many of the trials for breast, pancreatic and other cancer trials has been a contributing source. There will also be some major breakthrough news on stem cells but the emphasis in this issue will be turmeric's curcumin for fighting all inflammatory diseases.

DISEASES IN ISSUE 8

Interview with Dr. Carlos Lima who has performed 150 successful spinal cord stem cell surgeries. Turmeric for heart disease, Stem cells for the heart, Women and Turmeric, a 4 heart attack patient (renewed with stem cells) interviewed. How Stem cells will favorably impact health costs.

DISEASESIN ISSUE 9 (issues 9-12)

Stem cells for face lifts. Single cell grows prostate gland. Stem cells for baldness also stem cells for ALS. Amit Patel, M.D., Director Cardiovascular and Stem Cell Researchat University of Utah performs first time procedure and saves Coast to Coast listener who had 7 heart attacks. Alzheimer's and dementia discussion of the 7 stages and what is available for the fight. Also what is normal aging behavior and what is a warning sign. Much more!

DISEASES IN ISSUE 10

FIRSTstem cell trial for degenerative back disease. First trial for stroke victim recovery. Turmeric for breast cancer. Stem cells for cartilage. Turmeric for liver and prostate. Stem cells for newborns of drug addicted mothers. Stem cells for Parkinson's disease.

DISEASES IN ISSUE 11

HIV patient cured of AIDS,Stem Cells for Vision, Stem Cells Building Lungs,Turmeric for better Chemo results, Breakthroughs for Parkinson's, Building body parts, Stem Cells for deadly infections AND MORE.

DISEASES IN ISSUE 12

Three stem cell trials for eyes, insulin from cord blood, Teeth Stem Cells may heal brain, New MS study, Melanoma breakthrough, Stem Cells double muscle size for elderly, Progress report on Stem Cell research.

DISEASES IN ISSUE 13

Major cancer breakthrough, Stem Cells for brain cancer, Turmeric reduces stroke damage, Salute to a stem cell Pioneer, Dr. Amit Patel, Stem Cell Spray for Burn Victims using stem cells. Progressive MS Study, Stem Cells for family pets. Fewer aputations for diabetics, Fat Cells for breast reconstruction, Big Pharma making drugs from turmeric.

DISEASES IN ISSUE 14

Storing new borns umbilical cords for stem cells, Creating actual body parts in regenerative medicine, A 1st time reversal of Cystic Fibrosis, Stem Cells save Pro Pitcher's Arm, more on MS, Arthritis and joint damage repair, Kidneys and livers, Bone cartilage and joints, Correcting blindness in infants. Vatica financing adult stem cell research.

DISEASES IN ISSUE 15

Stem Cell war against 23 autoimmune diseases, awarded 1 of the top ten breakthroughs of the decade. Top athletes like Peyton Manning, Jason Kidd healing sports injuries with adult stem cells. Turmeric for brain cancers and gliomas. Stem cells building human body parts now. Dental Pulp to heal brains? Turmeric for kidneys.

DISEASES IN ISSUE 16

Stem Cell progress, Where are we? Sir Ian Wilmut cloned Dolly The Sheep now favors (iPSCs)Induced Pluripotent stem cells as the best answer for cures. First embryonic stem cell terminated because of tumors. Stem Cell hope to end Tissue Rejection. Most promising back stem cell trial, can it help you?

THE QUARTERLY STEM CELL REPORTMAY BE ORDERED ONLINE BY CLICKINGON THE ABOVE COLORED IMAGE

OR BY SENDING A CHECK OR MONEY ORDER FOR ONE YEAR FOR$19.95 ALONG WITH $2.50 S/H. TOTAL ONEYEAR PRICE IS $22.45 CA RESIDENTS KINDLY ADD $1.89 CA TX .

TO WILDE RESEARCH REPORT, P.O. BOX 1363, STUDIO CITY, CA 91614.You will receive all issues you order at one time.

SPECIFY WHICHCOMBINATION OF 4 ISSUESYOU WOULD LIKE --All16 issues are $79.80.

ORDERING OPTION, YOU MAY ALSO ORDER BY CALLING TOLL FREE, 1 866 STEM 123 AND LEAVE A MESSAGE FOR AN OFFICE CALL BACK TO TAKE YOUR ORDER.

* STEM CELLS FOR PETS*

YOUR dogs, cats and horsescancurrentlyreceive treatment with stem cells! Thesetreatments are not subject to FDA trial approval and are available now! The report will share information on how to contact the company that will direct you to veterinarians in your area who are certified and trained to perform stem cell treatment for your pet. Hip dysplasia, osteoarthritis, bone/ ligament disease,kidney and liverdisease (as examples) may be helped.

Disclaimer: The information presented in the newsletter is not to be misconstrued as endorsement or recommendation by the author or the reportfor any trial or therapy. It is acknowledged, many of these treatments are experimental and exploratory, therefore the information is presented to inform you as to what is being studied within the research communitythat might favorably impact yourcondition.Yourphysicianis your best arbiter andit is advisable to alwaysconsult with him or heron all decisions particularly with exploratory treatments.

Sincerely yours, Christian Wilde

Christian Wilde, "Hidden Causes of Heart Attack" and "Mir.

Go here to read the rest:
Stem Cell Heart Regeneration - Turmeric Curcumin Benefits ...

Read More...

stem cell therapy could help cats with kidney disease …

Wednesday, November 23rd, 2016

Most cat lovers have been touched by kidney disease at least once in their life. I lost my beloved Freddie at age 15 to this silent killer. A new procedure using adult stem cells to facilitate kidney transplantation in cats is being pioneered by the University of Georgia College of Veterinary Medicine.

The treatment of kidney failure in cats has traditionally been limited to changing diet, fluid therapy and a variety of medications and nutritional supplements. In the best cases, we can extend the life of affected cats by a handful of years if diagnosed early.

About 17,000 humans undergo kidney transplantation each year in the US and many enjoy a normal life expectancy after receiving their new kidney. In comparison, only a few cats undergo kidney transplant each year at only three transplant programs based at veterinary teaching hospitals. The low number of feline kidney transplants is primarily due to high cost, organ rejection and complications and ethical dilemmas involving the donor cat.

Cost and ethics aside, many cats are deemed poor transplant candidates. By the time kidney transplant is considered, the cat is often too ill or has developed too many complications. Organ rejection is a primary concern for many of these debilitated patients.

Researchers at the University of Georgia are pioneering the use of adult or mesenchymal stem cells (MSCs) to lower the risk of organ rejection in cats, especially those at higher risk for organ rejection. This procedure is being used for the first time in feline patients after a 2012 study of humans patients. The study found those receiving adult stem cells in conjunction with kidney transplantation had lower risk of organ rejection, fewer post-operative infections and better kidney function one year later.

It looks like adult stem cells help cats in the same ways. To date, two cats have undergone the procedure and are doing incredibly well. Adult stem cells in the UGA cases were obtained from fat tissues and then grown in a lab for about ten days before surgery. According to the researchers, stem cells used without kidney transplantation hasnt shown much success so far in treating chronic renal disease. Other cat candidates are currently being considered for this groundbreaking procedure.

Of course, this procedure is still quite expensive. From an ethical perspective, families of a cat that receive a donated kidney are required to adopt the donor cat, pledge to care for the donor cat for life and commit to treating both the recipient and donor cats.

Most recipient cats will require lifelong medications and injections, often twice a day, to prevent organ rejection. Stem cell therapy doesnt eliminate anti-rejection medication. Stem-cell treatments have been used with some success in treating certain musculo-skeletal conditions, but long-term studies are lacking.

Kidney disease is one of the most common causes of death in cats. I welcome any advances in battling this devastating condition. I understand that kidney transplantation may not be appropriate or possible for the majority of my patients. I appreciate these high-tech advances because I know they represent future breakthroughs that will benefit my typical patients.

If your cat is drinking more water, urinating more frequently, or inexplicably losing weight, have her checked by your vet immediately. Early diagnosis is still our best hope for extending the longevity and quality of life for cats enduring kidney failure.

See the article here:
stem cell therapy could help cats with kidney disease ...

Read More...

Stem cell therapy for heart failure: first implant of …

Thursday, August 4th, 2016

16.01.2015 - Press release

Circulation, metabolism, nutrition

On the 21 October 2014, Professor Philippe Menasch and his team from the cardiovascular surgery service of the Georges Pompidou European Hospital, AP-HP, carried out a transplant of cardiac cells derived from human embryonic stem cells*, according to a method developed by the Department of Cell and Tissue Biotherapies of the Saint-Louis hospital, directed by Professor Jrme Larghero and through research led by this group within Inserm. The surgery, coupled with a coronary bypass*, was carried out on a woman of 68 years suffering from severe heart failure. Ten weeks after the intervention, the patient is feeling well, her condition has improved markedly, with no complications having been observed. This promising advance was presented this Friday, 16 January 2015 at the XXV European Days Conference of the French Society of Cardiology.

Human embryonic stem cells. Transplantation of undifferentiated human embryonic stem cells into rat heart organotypic cultures. Presence of human cells, in the cardiac parenchyma of the rat two months after injection. The human cells are positive for human nuclear antigen marking (red). Cardiac rat tissue is positive for cardiac troponin 1 marking (green). I-Stem (Institiute for Stem Cell Therapy), Evry Genopole. Inserm/Habeler, Walter

The transplant was carried out as part of a clinical trial developed by the Public Hospitals of Paris (AP-HP) and through the work of the teams from AP-HP, Inserm and the universities of Paris-Descartes and Paris-Diderot. The cardiac cells were prepared according to a technique developed by the Department of Cell and Tissue Biotherapies of the Saint-Louis hospital. The cytogenetics laboratory of the Antoine Bclre Hospital and the French General Agency for Health Products and Equipment also contributed to the preparation of this phase I trial which will enable the verification of the safety and feasibility of the procedure

For 20 years Professor Menasch, currently co-director of an Inserm team within PARCC (Paris Centre for Cardiovascular Research), and his colleagues have been involved in stem cell* therapy for heart failure.

The team first tested the implant of skeletal muscle stem cells in necrosed areas of the heart in the laboratory. These cells were implanted into the heart of a patient with heart failure for the first time in the world on 15 June 2000. Following an initial series of these implants, always coupled with a coronary bypass, the team coordinated a European multi-centre, randomised, placebo-controlled trial whose results have still not been able to establish any significant benefit of these cells on the contractile function of patients hearts. One of the conclusions drawn from this trial was that to be fully efficient, transplanted cells should resemble the cells of the tissue to be repaired as much as possible, in this instance cardiac tissue. It was then decided to venture along the path of embryonic stem cells. Derived from embryos conceived in in vitro fertilisation, these cells do in fact possess pluripotent properties, that is, they are capable of developing into any type of cell of the body, including of course cardiac cells, as soon as they receive the appropriate signals during the culture cycle in the laboratory.

In 2007, the team then composed of, among others, Michel Pucat, Director of Research at Inserm, and Philippe Menasch showed that human embryonic stem cells could be differentiated into cardiac cells after being transplanted into the failing hearts of rats. Since then, many experiments have been carried out on different animal species in order to validate the efficacity of these cells and to optimise conditions which can guarantee maximum safety. At the end of this stage, a bank of pluripotent embryonic stem cells was formed in the conditions which satisfied all regulatory constraints applying to biological products for human use. Then, the Department of Cell and Tissue Biotherapies of the Saint-Louis hospital, still in liaison with the Inserm teams, developed and tested specialisation procedures for cells in order to produce young cardiac cells from them. The focus was then on the purification of the cells directed like so in order to ensure that the final product was expunged of any remaining pluripotent cells which could be potentially tumorigenic.

Besides, as initial experience with muscular stem cells showed the limitations of administering cells by multiple injections, their transfer is now performed using a patch that the cells are incorporated into. This patch is then placed on the area of the infarction. To that end, after the purification stage, the cardiac cells are incorporated into a circular fibrin gel which is applied, during the surgical procedure, to the necrosed area with just a few sutures ensuring that it is anchored to the cardiac tissue.

This type of surgery is aimed at serious heart failure which doesnt respond to the usual medicinal treatments but is not at the stage of a complete heart transplant. This is a promising advance, which we hope will enrich the therapeutic arsenal available to treat heart failure today explains Prof. Menasch. We are continuing the trial, which authorises us to carry out four other transplants. It would seem already that the benefits of the cells are linked mainly to the substances that they secrete. The direct administration of the substances, without going through a transplant of productive cells, is a path to explore.

This project has been entirely financed by funds from public intstitutions and societies and was authorised by the French National Agency for the Safety of Medicines and Health Products (ANSM) after agreement with the Agency for Biomedicine for the importation and research on human embryonic cells.

Cell therapy: refers to cell transplants aiming to restore the function of tissue or an organ when it has been altered by an accident, illness or ageing. These therapies have benefited from recent scientific advances on stem cells and give millions of patients the hope of regenerative medicine.

Embryonic or pluripotent stem cells: they can renew indefinitely (self-renewal), multiply in a culture and be differentiated into more than 200 types of cell. In the course of development, they are destined to form all types of the bodys tissue.

Coronary bypass: a technique that enables the redirection of the bloodstream towards the cardiac muscle, by using a graft (coming from the saphenous vein or from a thoracic artery.) One end of the graft is connected to the aorta, the main artery supplying the coronary arteries; the other end is connected to the coronary artery, situated just behind the site of the obstruction. This creates a detour enabling the oxygenated blood to circulate towards the heart.

Circulation, metabolism, nutrition

30.07.2014 - Press release

By combining fundamental research and monitoring a single cohort of kidney-transplant patients with antiphospholipid syndrome, the researchers have highlighted a beneficial effect of sirolimus, commonly used as an immunosuppressor in organ transplants, to prevent recurrence of vascular lesions on the transplanted kidney. ...

24.09.2015 - Press release

Des scientifiques provenant de 6 pays, runis au sein du projet ENS@T-HT mettent en commun leur expertise afin damliorer le diagnostic et la prise en charge thrapeutique de lhypertension artrielle primaire et secondaire par une approche axe sur les omiques . ...

07.10.2014 - Video press release

Eating disorders (ED) such as anorexia nervosa, bulimia, and binge eating disorder affect approximately 5-10% of the general population, but the biological mechanisms involved are unknown. Researchers at Inserm Unit 1073, Nutrition, inflammation and dysfunction of the gut-brain axis (Inserm/University of Rouen) ...

10.02.2014 - Press release

European cutting-edge research will be attended at the annual meeting of the American Association for the Advancement of Science (AAAS) in Chicago. ...

Follow this link:
Stem cell therapy for heart failure: first implant of ...

Read More...

Stem Cells to be Tested for Kidney Repair

Thursday, August 4th, 2016

hidden

Salt Lake researchers are launching a groundbreaking clinical trial to see if adult stem cell transplants will reverse or prevent kidney failure. If it works, it will be the kind of self-healing everybody has been waiting for. Stem cell transplants have proven successful in animal experiments in Germany and Salt Lake, but now the time has come to start clinical trials in humans. Two patients here have already had the stem cell transplants. Open heart surgery places a lot of stress on the kidneys, patients who have other complications often go into kidney failure. That's why this group has been selected for the clinical trial. A special kind of adult stem cell taken from the bone marrow of living donors will be injected into the blood stream shortly after their heart surgeries. Christof Westenfelder, M.D., the Chief Medical Officer at Allocure, said "These cells, after they read what's going on in the injured organ, then instruct the surviving cells in the injured organ to defend themse

Salt Lake researchers are launching a groundbreaking clinical trial to see if adult stem cell transplants will reverse or prevent kidney failure. If it works, it will be the kind of self-healing everybody has been waiting for. Stem cell transplants have proven successful in animal experiments in Germany and Salt Lake, but now the time has come to start clinical trials in humans. Two patients here have already had the stem cell transplants. Open heart surgery places a lot of stress on the kidneys, patients who have other complications often go into kidney failure. That's why this group has been selected for the clinical trial. A special kind of adult stem cell taken from the bone marrow of living donors will be injected into the blood stream shortly after their heart surgeries. Christof Westenfelder, M.D., the Chief Medical Officer at Allocure, said "These cells, after they read what's going on in the injured organ, then instruct the surviving cells in the injured organ to defend themselves, to repair the organ." The stem cells linger until the repair is complete, then, as programmed, self-destruct within three days so they won't go to other organs where they're not needed.

View Full Story

The rest is here:
Stem Cells to be Tested for Kidney Repair

Read More...

Stem Cell Therapy for Kidney Failure Treatment of CKD,CRF …

Thursday, August 4th, 2016

Mrs Dily from Papua New Guinea talks about her Hematopoietic Cord Blood Mesenchymal stem cell treatment for Kidney Failure More here https://stemcellthailand.org/therapie...

The condition was due to Diabetes Type 2 complications and required nephrogenesis therapy.

The Regeneration Center of Thailand provides the most advanced medical therapy for Renal Failure using non-invasive Renal tube regeneration therapy to naturally treat Organ Dysfunctions including Kidney cell replacement therapy for patients diagnosed with Kidney disease and End Stage Renal Failure CKD CRF and offers and alternative to having kidney transplants.

Human kidneys normally have the capacity to regenerate themselves and restore their normal function and structure after most injuries, however, kidney regeneration is usually delayed or combine with multiple conditions resulting in gradual decrease failure and mortality. Incomplete natural regeneration of kidneys are especially exposed for Diabetics and often lead to loss of renal function. Our kidneys normally undergo mesenchymal-epithelial development stage of life and an adult kidney contains about 24 types of cells that are arranged in distinct vascular, glomerular, interstitial and tubular compartments.

Our enriched Hematopoietic Mesenchymal cell therapy and nephrogenesis protocol works buy enhancing our bodies natural defences against kidney damage and look to reconstitute a failing or injured kidney by improving both the structural compartments and their functions.

Enriched Renal Stem cells help to repair injured kidneys by replacing the dead cells and/or by augmenting natural regeneration of the infused cells through the paracrine effect. Cell-based treatments for Renal failure also require proper lab expansion of very large cell populations that must be uniform in their activity and be tested to ensure they are pathogen-free before delivery to the patient.

The statistics for people suffering from ARF and acute renal failure across the world are not pleasant. Nearly 7% of all long-term hospital patients across the world are suffering from Kidney failure and the mortality rate for these patients is about 50% and has basically remained unchanged since the invention of dialysis nearly 40 years ago.

The main strength of renal stem cell transplants involves the introduction of enriched supplementary cells into a damaged kidneys to more effectively aid in the repair and regeneration that are experience in the natural healing process.

Stem Cell transplants for Kidney failure and Chronic Kidney disease are not appropriate for all patients. To see if you qualify for treatment please contact us below.

EMAIL:info@stemcellthailand.org SKYPE: StemCell.Regen

FACEBOOK: http://facebook.com/stemcellsthailand GOOGLE+: http://google.com/+StemcellthailandOrg

PHONE NUMBERS Australia: 02-8006-1094 New Zealand: 09-889 3777 USA and Canada: 1-347-450-THAI (8424) London: 20 3286 1444 Thailand +66 808 069 391

ADDRESS Regeneration Center of Thailand 808/8 Thararom 2 Sukhumvit 55 FL 2 Bangkok 10110 Thailand

The Regeneration Center of Thailand 2015

See the original post here:
Stem Cell Therapy for Kidney Failure Treatment of CKD,CRF ...

Read More...

Kidney Transplant | Stem Cell Foundation

Thursday, August 4th, 2016

Kidney transplant (also called renal transplant) is the placement of a donor kidney into a patient with end-stage renal disease (ESRD).Kidney transplants are classified as deceased-donor (formerly called cadaveric donor) or living-donor transplants depending on the source of the donor organ. Living-donor renal transplants are further characterized as genetically related (living-related) or non-related (living-unrelated) transplants, depending on whether a biological relationship exists between the donor and recipient.

The first diseased-donor kidney transplant in the United States was performed in 1950 on Ruth Tucker, a 44-year-old woman with polycystic kidney disease (PKD), at Little Company of Mary Hospitalin Evergreen Park, Illinois. Although the donated kidney was rejected ten months later because no immunosuppressive therapy (anti-rejection medication)was available at the time (the development of effective anti-rejection drugs was years away), the intervening time gave Tuckers remaining kidney time to recover and she lived another five years.

The first kidney transplant between living patients was undertaken in 1954 in Boston and Paris. The Boston transplant, performed on December 23, 1954, at Brigham Hospital was performed by Joseph Murray, J. Hartwell Harrison, John P. Merrill and others.The procedure was done between identical twinsto eliminate any problems from an immunereaction. For this and later work, Dr. Murray received the Nobel Prize for Medicinein 1990. The recipient lived for eight years after the transplant.

The first kidney transplantation in the United Kingdom occurred in 1960, when Michael Woodruffperformed one between identical twins in Edinburgh.Until the 1964 introduction of anti-rejection medications to prevent and treat acute rejection, deceased-donor transplants were not performed.

Kidney was the easiest organ to transplant: tissue typing was simple, the organ was relatively easy to remove and implant, live donors could be used without difficulty and in the event of failure, kidney dialysiswas available (dialysis had been in use since the 1940s).

The development of increasingly effective immunosuppressive therapies has increased the average life of a transplanted kidney to about 20 years, after which the recipient may be considered for a second transplant or require regular dialysis. Anti-rejection drugs suppress the recipientss immune system to keep it from attacking the transplanted organ as an invader, and must be taken for life to prevent rejection. Suppressing the immune system long term, however, makes the recipient vulnerable to infections and cancers that would not otherwise be a problem. In addition, the drugs themselves have side effects ranging from osteoporosis, appearance changes, cardiovascular disease and kidney damage. The cost of drugs and treatment generally run between $25,000 and $45,000 per year for the life of the patient.

The indication for kidney transplantation is end-stage renal disease(ESRD), regardless of the primary cause.Diabetes is the most common cause of kidney transplantation, accounting for approximately 25% of transplants in the U.S. The majority of renal transplant recipients are on some form of dialysis atthe time of transplantation. Individuals with chronic renal failure who have a living donor available, however, may undergo pre-emptive transplantation before dialysis is needed.

Common diseases leading to ESRD include:

Find clinical trials here: Kidney transplant clinical trials

Read the rest here:
Kidney Transplant | Stem Cell Foundation

Read More...

Stem Cell Treat Kidney Failure – Kidneyabc.com

Thursday, August 4th, 2016

Quite a number of people throughout the world are tormented by kidney disease, either inherited or acquired. Impaired kidneys make them suffer from various discomforts and complications. For this reason, every patient with kidney disease try to find a kidney rebuilding treatment. Stem cell therapy just gives a new hope for these patients.

Stem cells are a class of multipotential cells that are able to differentiate into various cells. According to developmental stages, stem cells are divided into embryonic stem cell (ES) and adult stem cells.

ES, also called almighty stem cells, can differentiate into all types of tissues and organs, while adult stem cells can only differentiate into several or some certain tissue or organ. Therefore, ES is better as the source of stem cell therapy.

Stem cell therapy refers to stem cells go to damaged areas and regenerate new cells and tissues in an appropriate condition. In recent years, this therapy has been used widely to treat respiratory diseases, cerebropathy, blood disease, liver diseases, etc. For kidney disease patients, stem cell therapy shows a quite potential effect on repairing damaged kidney inherent cells and rebuilding kidney normal structure.

Within normal kidneys, there are five types of renal intrinsic cells that guarantee kidneys to work perfectly, including: glomerular epithelial cells, glomerular endothelial cells, interstitial fibroblast, glomerular mesangial cells and renal tubular epithelial cells.

Kidney disease is a condition in which these renal intrinsic cells are attacked and lose their ability gradually, so kidney function, also known as GFR, declines accordingly. When kidneys can't work normally, more and more waste products and toxins build up in the body to cause various complications.

CKD, IgA Nephropathy, Kidney Failure, PKD, Lupus Nephritis, FSGS, Nephrotic Syndrome, Diabetic Nephropathy, Hypertensive Nephropathy, etc are all common types of kidney disease that affects patients' health largely.

In one sterile environment, a lot of stem cells are injected to patients' body. Then, they can go to damaged kidneys to differentiate into kidney inherent cells. For example, if glomerular epithelial cells are damaged, stem cells will differentiate into this kind of cells.

Once these new cells can play their work normally, it means kidney disease is treated fundamentally. Generally speaking, kidney disease patients may get the following benefits from this therapy:

- Repair damaged kidney cells and regenerate new cells

- Rebuild immune system through inhibiting the proliferation of T cells and immune reactions

- Improve kidney function largely

- Alleviate complications such as high creatinine level and hypertension

- Prevent the relapse of kidney disease

- Delay or even avoid dialysis

Even though stem cell therapy shows an obvious and irreplaceable effect on treating kidney disease, not every country has corresponding centers to do this therapy. Besides, not all kidney disease patients can receive stem cell therapy. If you still have some kidney function, you can consult the online doctor to ask the nearest hospital or center to get stem cell therapy or whether you can use this therapy.

View original post here:
Stem Cell Treat Kidney Failure - Kidneyabc.com

Read More...

Stem Cell Therapy in 2015 for Patients with Kidney Failure

Thursday, August 4th, 2016

Home > Understanding Kidney Disease > Kidney Diseases > Kidney Failure > Kidney Failure Treatment > 2015-07-21 16:16| Font Size A A A

Along with the development of medical technology, Stem Cell Therapy has been widely used to control or even stop progression of illness condition for patients with Kidney Failure. Here we will introduce one of most popular kidney disease treatment in 2015--Newest Stem Cell Transplantation Therapy.

What is the treatment?

Stem cell therapy is also known as stem cell transplant which has tremendous promise to help us understand and treat a range of diseases. Stem cell is primitive cell that has the potential ability of self replication and multi-directional differentiation, that is to say, stem cell is the origin of the body. The therapy uses healthy stem cells to transplant into the patients, so as to repair damaged cells and rebuild organ structure.

How does it treat patients with renal failure?

Actually, in Shijiazhuang Kidney Disease Hospital-one of largest kidney disease hospitals in China, we make full use of abilities of stem cells like self-renewing, high proliferation and multiple-directional differentiation to create the completely new, normal or even much younger cells, tissues and organs. When stem cells are transplanted into the body, these new and younger cells will find and replace the damaged or necrotic cells and tissues. In the condition, the treatment can help improve renal function from the root for patients with different type of kidney diseases like Kidney Failure, IgA Nephropathy, Lupus Nephritis, Diabetic Nephrpathy and so on.

What are the characteristics of the treatment?

According to clinical practice, we find the treatment has the following features:

- Safety: nontoxicity

- This therapy can be used, even though the pathogenesis of the disease hasnt been detected fully

- For some symptoms, stem cell transplant has an obvious effect.

- The source of transplanted stem cells is sufficient.

- Stem cells are the best vectors of immune therapy and gene therapy.

- It brings brand new hope to treat incurable diseases.

If you still have other questions or have interest in the therapy, please contact with our Online Doctor or email to us at renal-disease@hotmail.com and we will do our best to help you.

Read this article:
Stem Cell Therapy in 2015 for Patients with Kidney Failure

Read More...

Kidney disease following hematopoietic cell transplantation

Thursday, August 4th, 2016

OVERVIEW

Hematopoietic cell transplantation (HCT) is the only cure for a variety of hematologic and oncologic diseases. However, it has been associated with the development of both acute and chronic kidney failure [1]. (See "Preparative regimens for hematopoietic cell transplantation".)

The term "hematopoietic cell transplantation" will be used throughout this review as a general term to cover transplantation of progenitor cells from any source (eg, bone marrow, peripheral blood, cord blood). Otherwise, the source of such cells will be specified (eg, autologous peripheral blood progenitor cell transplantation). (See "Sources of hematopoietic stem cells".)

Although the requirement for dialysis following HCT is relatively uncommon (in the range of 2 to 5 percent), it is generally associated with an extremely poor prognosis. However, acute kidney injury (AKI), as defined by doubling or smaller increases in the serum creatinine concentration, is common, approaching 50 to 60 percent in several reports. There are a variety of causes of AKI following HCT and several risk factors that are associated with chronic kidney disease (CKD). In the majority of patients, renal dysfunction is temporary and returns to baseline; however, adjusting medications to avoid further damage is common.

This topic will review the epidemiology and causes of kidney injury following HCT other than sepsis or drug-induced acute tubular necrosis or tumor lysis syndrome. These include hepatic sinusoidal obstructive syndrome (SOS, formerly known as veno-occlusive disease), hemolytic-uremic syndrome/thrombotic microangiopathy, and calcineurin inhibitor nephrotoxicity. The technique and other complications associated with HCT are discussed separately. (See "Preparative regimens for hematopoietic cell transplantation" and "Hematopoietic support after hematopoietic cell transplantation".)

EPIDEMIOLOGY AND PROGNOSIS

The epidemiology and risk factors of kidney injury following hematopoietic cell transplantation (HCT) vary with the different types of regimens associated with HCT:

Literature review current through: Feb 2016. | This topic last updated: Wed Sep 09 00:00:00 GMT 2015.

Read this article:
Kidney disease following hematopoietic cell transplantation

Read More...

Kidney Disease Therapy Knowledge-Kidney Failure

Thursday, August 4th, 2016

What are the current treatment options for kidney diseases? Here is the overview of blood purification technology, kidney transplant, Chinese medicines (acupuncture, cupping therapy, Gua Sha, Qi Gong, massage, Taijiquan, pedicure, medicated bath) and the latest immunotherapy. We will introduce you their adaptable diseases, advantages and disadvantages. You can make the best choice according to your specific illness and physical conditions. Of cause you should consult the doctors for advise and guidance.

Dialysis is to purify the blood and treat renal failure.

Hemodialysis can help remove wastes and sustain life.

Peritoneal dialysis is to use peritoneum as semipermeable membrane.

Plasma exchange uses fresh plasma to replace toxic materials.

Immune adsorption is to use adsorbent to remove pathological factors

Blood perfusion is often combined with hemodialysis in clinic.

Hemofiltration is the exchange of ultrafiltrate and substitute.

Blood pollution treatment is a brand new treatment for kidney disease.

Acupuncture is to use needles to activate meridians and acupuncture points.

Cupping therapy is to use pots to cause congestion or blood stasis.

Gual Sha is to use jade or horn to rub certain parts of the skin.

Massage is friction, knead or knocking of the surface of body.

Qigong is regulation of respiration, physical and mental activity.

Taijiquan is one type of Chinese Kong fu for building up physical strength.

Medicated bath is to treat disease by taking bath with medicines in water.

Pedicure is to stimulate the reflection zone in the feet.

The rest is here:
Kidney Disease Therapy Knowledge-Kidney Failure

Read More...

Rob Waddell Defeats Kidney Disease – Adult Stem Cells

Thursday, August 4th, 2016

Rob Waddell Defeats Kidney Disease

Rob Waddell knew at an early age that he would need a kidney transplant. His mother has had two transplants and polycystic kidney disease runs in his family. "Ive had two uncles thatve died from this disease. At early ages. I mean they went on dialysis, they had a transplant, something happened, theyre no longer here. Their kids are, without, without a dad!", Rob said.

So when his doctor told him he had to go on dialysis and that a transplant was imminent it was no surprise. Having watched his mother suffer the ups and downs of taking anti-rejection drugs her whole life, he was thrilled to find out there was another option. He entered a clinical trial whereby he would receive an adult stem cell transplant from his kidney donor at the time of the kidney transplant surgery. The donors adult stem cells would allow Rob to accept the same donors kidney, essentially re-training his immune system so that it would recognize the donor kidney as part of Robs own body.

Rob says, "Well, I decided to do the stem cell transplant because I didnt want to live the rest of my life on immune rejection drugs. The good and the bad of immune suppressant drugs is they let the kidney stay in your body. The bad part is that slowly over time it kills the kidney. Its toxic to the kidney. So those drugs, over time, will cause the kidney to fail. My wife, Karen, she, when I proposed the idea of me doing this stem cell study, she was really kind of concerned. I mean she didnt want me to do it because it was new."

Karen Waddell remembers what she said when she heard about the adult stem cell transplant, "I told him I was totally against it from the beginning. Didnt like it. I said, you can just have a normal transplant. Your mom has lived through it. You know, well just adjust."

Rob says, "Seeing my mother go through the repercussions of having kidney disease and the transplant and immune rejection drugs, probably was the number one foundation for me pursuing this."

After the stem cell infusion, Karen says, Rob was like a new man. "Its like hes rejuvenated. Its amazing. He's alert. All his faculties are working great. And for him to be just drug-free, oh its wonderful!

"We call him my fifth child and other people that know us too, theyll tease, because you will see him rip-sticking around the neighborhood, or on the trampoline. So Im thankful that he was able to just be determined and have that drive and the foresight to know that he was going to get those stem cells.

Today Rob lives a full and active life chasing four kids around the soccer, baseball and lacrosse fields of Louisville, Kentucky.

"I feel so fortunate, because Ive been blessed with this. I mean truly a new lease on life. I feel fantastic. My kids could tell you that. I mean I wear them out half the time and I didnt before.

Actually, almost every day since then, I just walk around and Im like, Wow! I feel so goodI mean is this really happening? These adult stem cells to me were a chance to live a normal life.and its amazing."

See the original post here:
Rob Waddell Defeats Kidney Disease - Adult Stem Cells

Read More...

Mesenchymal stem cell treatment for chronic renal failure …

Thursday, August 4th, 2016

Chronic renal failure is an important clinical problem with significant socioeconomic impact worldwide. Despite advances in renal replacement therapies and organ transplantation, poor quality of life for dialysis patients and long transplant waiting lists remain major concerns for nephrologists treating this condition. There is therefore a pressing need for novel therapies to promote renal cellular repair and tissue remodeling. Over the past decade, advances in the field of regenerative medicine allowed development of cell therapies suitable for kidney repair. Mesenchymal stem cells (MSCs) are undifferentiated cells that possess immunomodulatory and tissue trophic properties and the ability to differentiate into multiple cell types. Studies in animal models of chronic renal failure have uncovered a unique potential of these cells for improving function and regenerating the damaged kidney. Nevertheless, several limitations pertaining to inadequate engraftment, difficulty to monitor, and untoward effects of MSCs remain to be addressed. Adverse effects observed following intravascular administration of MSCs include immune rejection, adipogenic differentiation, malignant transformation, and prothrombotic events. Nonetheless, most studies indicate a remarkable capability of MSCs to achieve kidney repair. This review summarizes the regenerative potential of MSCs to provide functional recovery from renal failure, focusing on their application and the current challenges facing clinical translation.

Chronic kidney disease (CKD) is a prevalent condition (8 to 16%) associated with all-cause and cardiovascular mortality [1]. Importantly, CKD can progress towards end-stage renal disease (ESRD), requiring renal replacement therapy. ESRD currently accounts for 6.3% of the Medicare spending in the United States, and is projected to increase by 85% by 2015 [2]. Furthermore, ESRD has a tremendous impact on quality of life and life expectancy of affected individuals [3]. Therefore, it is imperative to develop therapeutic interventions to prevent, alleviate, or decelerate progression of renal failure.

Diabetes mellitus and hypertension represent major causes of CKD and initiation of dialysis in the United States [4]. In addition, glomerular diseases, malnutrition, infectious diseases, and acute kidney injury can progress to ESRD, contributing to the increased global burden of death associated with this condition [5]. Current treatment modalities often fail to target the major underlying contributors for progression of renal disease [6]. Chronic glomerular and tubulointerstitial fibrosis is a common pathway to ESRD, often associated with apoptosis, oxidative damage, and microvascular rarefaction. In fact, renal dysfunction is postulated to better correlate with the degree of tubulointerstitial than with glomerular damage [7].

Importantly, the kidney possesses intrinsic regenerative capacity that allows the organ to recover after limited insults [8]. Unfortunately, this regenerative potential is limited under chronic conditions and thus inefficient to prevent progressive glomerulosclerosis and tubulointerstitial fibrosis [9]. Treatment strategies that boost cellular regeneration might therefore offer good alternatives for patients with CKD.

Mesenchymal stem cells (MSCs) can be isolated from a variety of tissues, differentiate into multiple cell lineages, and possess unique immunomodulatory properties that ameliorate inflammation and immune responses, constituting a promising tool to facilitate renal repair. MSCs are defined by the presence of plastic-adherent cells under standard culture conditions, capacity to differentiate into osteoblasts, adipocytes and chondroblasts in vitro, expression of typical surface markers (CD29, CD44, CD73, CD90, CD105, and CD166), and the lack of CD45, CD34, CD14 or CD11b, CD79 or CD19 and HLA-DR surface molecules [10]. In recent years, experimental studies have uncovered the potential of MSCs to improve renal function in several models of CKD, and several clinical studies have indicated their safety and efficacy in CKD. Nevertheless, a number of hurdles need to be addressed before clinical translation. This review summarizes the current state of MSC transplantation for CKD, focusing on their mechanisms of renal repair, complications, obstacles for clinical translation, and potential approaches to overcome them.

Over the past few years, MSCs have been successfully applied in experimental models of CKD such as diabetes, hypertension, and chronic allograft nephropathy (Table

). For example, a single intravenous dose of MSCs resulted in beta-pancreatic islet regeneration, prevented renal damage in streptozotocin-induced type 1 diabetes in C57BL/6 mice [

], and decreased hyperglycemia and glycosuria that persisted for 2months after injection. Furthermore, MSC-treated diabetic mice showed histologically normal glomeruli, and albuminuria fell. Although the authors did not assess cellular mechanisms associated with MSC therapeutic effects, the long-lasting persistence of injected MSCs may suggest a direct effect to elicit kidney regeneration.

Preclinical studies using mesenchymal stem cells for the treatment of chronic kidney disease

Diabetic nephropathy

Mice bone marrow

0.5106

Intravenous

Engraftment/direct effect

None

[11]

Diabetic nephropathy

Human bone marrow

2106

Intracardiac

Engraftment/direct effect

None

[12]

Partial nephrectomy

Rat bone marrow

1106

Intravenous

Paracrine effect

None

[13]

Chronic allograft nephropathy

Rat bone marrow

0.5106

Intravenous

Immunomodulatory effect

None

[14]

Renal revascularization

Allogeneic swine adipose tissue

10106

Intrarenal

Engraftment/direct effect/paracrine

None

[16, 17]

Renal artery stenosis

Autologous swine adipose tissue

10106

Intrarenal

Engraftment/direct effect/paracrine

None

[15]

Similarly, Lee and colleagues tested the effectiveness of intracardiac infusions of MSCs from human bone marrow in immunodeficient mice with type 2 diabetes produced with multiple low doses of streptozotocin [12]. MSCs lowered blood glucose levels and decreased mesangial thickening and macrophage infiltration, suggesting their potential for improving renal lesions in subjects with diabetes mellitus. Interestingly, in kidneys of MSC-treated diabetic mice, a few injected human MSCs differentiated into glomerular endothelial cells.

Additionally, in rats with modified 5/6 nephrectomy, a single venous injection of MSCs 1day after nephrectomy preserved renal function and attenuated renal injury [13]. Despite unchanged blood urea nitrogen and creatinine levels, MSC-treated animals showed attenuated progression of proteinuria. The scarce engraftment of MSCs in the kidneys of rats with chronic renal failure suggests that paracrine secretion of mediators, such as cytokines or growth factors, may have accounted for their beneficial effects. Indeed, vascular endothelial growth factor (VEGF) levels were substantially higher in MSC-treated animals 1month after MSC injection.

Furthermore, a single dose of bone marrow-derived MSCs 11weeks after kidney transplantation in rats decreased interstitial fibrosis, tubular atrophy, T-cell and macrophage infiltration, and the expression of inflammatory cytokines [14]. Interestingly, a decrease over time in the inflammatory and profibrotic cytokine levels in MSC-treated animals was associated with an increase in the anti-inflammatory cytokine IL-10, although none of the injected MSCs were detected 7days after delivery. These observations imply that the beneficial effect of these cells in this setting is primarily attributable to their paracrine immunomodulatory properties rather than long-term engraftment.

We have previously shown in swine atherosclerotic renovascular disease that intrarenal delivery of MSCs isolated from subcutaneous adipose tissue protected the stenotic kidney despite sustained hypertension [

]. Notably, MSCs also attenuated renal inflammation, endoplasmic-reticulum stress, and apoptosis through mechanisms involving cell contact. Furthermore, adjunctive MSCs improved renal function and structure after renal revascularization and reduced inflammation, oxidative stress, apoptosis, microvascular remodeling, and fibrosis in the stenotic kidney [

] (Figure

). This strategy also restores oxygen-dependent tubular function in the stenotic-kidney medulla, extending their value to preserving medullary structure and function in chronic ischemic conditions [

].

Stenotic-kidney microvascular loss and fibrosis decreased in animals treated with mesenchymal stem cells. Top: representative microcomputed tomography three-dimensional images of kidney segments, showing improved microvascular architecture in pigs with atherosclerotic renal artery stenosis (ARAS) treated with percutaneous transluminal renal angioplasty (PTRA) and an adjunct intrarenal infusion of adipose tissue-derived mesenchymal stem cells (MSC) 4weeks earlier. Bottom: representative renal trichrome staining (40, blue) showing decreased fibrosis in ARAS + PTRA + MSC pigs.

While preclinical studies have established the safety and efficacy of MSCs in different models of CKD, these results need cautious translation into routine clinical practice. Trials using MSCs for CKD patients may face various challenges, including selecting the optimal route of MSC delivery, scant homing and engraftment, immune rejection, ensuring thriving, and tracking of injected cells. Addressing these challenges may bolster the success of MSC therapy in improving renal function in CKD patients.

The route of MSC delivery may influence the cells capacity to home and engraft the damaged tissue, and thereby their efficacy for renal repair. Commonly used experimental methods to deliver MSCs include systemic intravenous, intra-arterial, or intraparenchymal delivery. When intravenously delivered in normal SpragueDawley rats, the majority of MSCs are initially trapped in the lungs [18], but in nonhuman primates the cells distribute broadly into the kidneys, skin, lung, thymus, and liver with estimated levels of engraftment ranging from 0.1 to 2.7% [19]. In contrast, direct delivery of MSCs into the renal artery of an ischemic kidney is associated with retention rates of 10 to 15% [16, 17], although the normal swine kidney retains only around 4%, due to the low tonic release of injury signals. However, injection of human MSCs into the mouse abdominal aorta may lead to occlusion in the distal vasculature due to their relatively large cell size (16 to 53m), suggesting that this approach should be used cautiously [20]. Injections of MSCs into the renal parenchyma or their local subcapsular implantation confer renoprotective effects [21, 22], but are difficult to implement in the human injured kidney.

In experimental models of CKD, the optimal dose of MSCs is often empirical, with doses ranging from 0.5106 to 10106[11, 16]. Despite variability in dose regimens and route of delivery, the safety and beneficial effects of MSCs were consistent among studies. Nevertheless, the use of escalating doses is strongly recommended in clinical trials, and chronic adverse events should be evaluated prior to enrollment at the next dose level.

Circulating hematopoietic progenitor cells home to the damaged kidney by responding to injury signals that correspond to cognate surface receptors which they express [23]. Accumulating evidence indicates that exogenously infused MSCs respond to similar homing signals. In mice, expression of CD44 and its major ligand hyaluronic acid mediates MSC migration to the injured kidney [24], and hyaluronic acid also promotes MSC dose-dependent migration in vitro. Moreover, renal homing of intravenously injected MSCs was blocked by preincubation with the CD44 blocking antibody or by soluble hyaluronic acid, suggesting that CD44 and hyaluronic acid interactions recruit exogenous MSCs to the injured kidney. In addition, Liu and colleagues found that, when administered systemically, MSCs home to the ischemic kidney, improving renal function, accelerating mitogenic response, and reducing cell apoptosis, but these effects were abolished by either CXCR4 or CXCR7 inhibition, implicating the stromal derived factor-1CXCR4/CXCR7 axis in kidney repair [25].

Collectively, these observations suggest that strategies aimed to enhance MSC expression of homing signals may improve their capacity to attenuate renal dysfunction. Studies have shown that selective manipulation of MSCs before transplantation (preconditioning) enhances their ability to protect damaged tissues [26, 27]. The rationale underpinning this approach is that transplanted MSCs encounter a hostile microenvironment that mitigates their reparative capabilities and survival. Indeed, preconditioning with the mitogenic and prosurvival factor insulin-like growth factor (IGF)-1 before systemic infusion of bone marrow-derived MSCs (2105) upregulates the expression of CXCR4 and restores normal renal function in a mice model of cisplatin-induced acute kidney injury [28].

Some studies suggest that MSCs have the capacity to engraft the damaged tissue, integrate into tubular cells, and differentiate into mesangial cells [2931]. In swine renovascular disease, 4weeks after intrarenal infusion, MSCs (10106) were detected in all regions of the kidney, but mostly at the renal interstitium [16, 17]. On the other hand, intravenous infusion of bone marrow-derived MSCs (2105) in mice with cisplatin-induced acute renal failure reduced the severity of renal injury, but none were detected within the renal tubules and only few cells within the renal interstitium at 1 to 4days after infusion [32], suggesting that MSC engraftment is not necessary to achieve renoprotection. Likewise, despite significant improvement in renal function, within 3days of intracarotid infusion in a rat model of ischemiareperfusion-induced acute renal failure, none of the MSCs differentiated into the tubular or endothelial cell phenotype, indicating that their beneficial effects are primarily mediated via paracrine actions rather than differentiation into target cells [33].

Methods to increase MSC engraftment may therefore enhance their utility in regenerative cellular therapy. Temporary obstruction of the renal artery following intrarenal delivery [16, 17] may prevent cell washout, and is associated with significant retention rates in the postischemic kidney. Alternatively, in a rat model of acute kidney injury, s-nitroso N-acetyl penicillamine preconditioning enhances MSC engraftment, ultimately associated with a significant improvement in renal function [34].

Despite the crucial role attributed to MSC engraftment in potentiating the cells beneficial effect at the site of injury, there is currently consensus that the chief mechanism by which MSCs protect the damaged kidney is the release of growth factors, proangiogenic factors, and anti-inflammatory cytokines. Cultured MSCs release large amounts of the proangiogenic factor VEGF, which facilitates glomerular and tubular recovery [16, 35]. MSCs can also produce IGF-1, while administration of IGF-1 gene-silenced MSCs limits their protective effect on renal function and tubular structure in murine cisplatin-induced kidney injury, indicating that MSCs exert their beneficial effects by producing IGF-1 [36].

Importantly, these paracrine actions of MSCs seem to mediate their immunomodulatory properties. In ischemiareperfusion-induced acute kidney injury, infusion of MSCs downregulates renal expression of proinflammatory cytokines and adhesion molecules such as IL-1, tumor necrosis factor alpha, interferon gamma, monocyte chemoattractant protein-1, and intercellular adhesion molecule-1, but upregulates the expression of the anti-inflammatory IL-10 [26, 33]. Likewise, we have shown in swine renovascular disease that intrarenal delivery of MSCs during renal revascularization decreased renal expression of tumor necrosis factor alpha and monocyte chemoattractant protein-1, but increased IL-10 expression [17]. Moreover, MSCs induced a shift in the macrophage phenotype from inflammatory (M1) to reparative (M2), uncovering their immunomodulatory potential [37]. Taken together, these observations underscore the contribution of paracrine actions of MSCs to induce a shift from an inflammatory to an anti-inflammatory microenvironment. It is not unlikely that the type, number, and expansion methods used to secure MSCs alter their engraftment capacity.

For many years, MSCs have been considered immune privileged because of the lack of expression of co-stimulatory molecules and their capacity to decrease renal release and expression of inflammatory mediators [17, 33, 37]. These attributes engendered the hope that MSCs could engraft in allogeneic nonimmunosuppressed recipients, and stimulated development of off-the-shelf allogeneic MSC products [38], which allow rapid generation of large amounts of cells from few donors. Nevertheless, in vivo and in vitro studies have demonstrated that MSCs may occasionally induce an immune switch transitioning from an immunoprivileged to an immunogenic phenotype that triggered cellular cytotoxicity or immune rejection [39]. Moreover, implantation of murine MSCs engineered to release erythropoietin in major histocompatibility complex-mismatched allogeneic mice increased the proportion of host-derived lymphoid CD8+ and natural killer infiltrating cells, suggesting that MSCs are not intrinsically immunoprivileged [40]. Taken together, these observations do not support the use of allogeneic MSCs as a universal cellular platform, at least until development of unequivocally immunoprivileged MSCs. Therefore, at this point, administration of autologous MSCs seems to be the safest strategy.

An important feature of MSCs is their capacity to induce proliferation of renal glomerular and tubular cells, increasing cellular survival. By secreting proangiogenic and trophic factors, injected MSCs not only can enhance proliferation, but also can decrease apoptosis of tubular cells [32]. We have shown in swine renovascular disease that a single intrarenal delivery of MSCs in conjunction with renal revascularization increased proliferation of renal cells [16], and recently confirmed in vitro that MSCs blunt apoptosis by decreasing the expression of caspase-3 [15].

However, whether MSCs remain in the circulation long enough to exert any long-lasting effect is a matter of debate. Ezquer and colleagues showed that intravenous MSCs home into the kidney of type 1 diabetic mice, and some donor MSCs remained in the kidney up to 2months later [11]. Similarly, we found that 4weeks after intrarenal delivery a significant number of MSCs were retained in the injected kidney [16, 17], whereas by 12weeks after cell transfer only a few cells were observed in the kidney, yet their beneficial effects were sustained [15]. Longitudinal studies are needed to document the chronology of MSC retention and beneficial benefits in the kidney. Additionally, development of novel interventions such as preconditioning may enhance survival and potency of MSCs in renal failure. For instance, MSCs exposed to hypoxic conditions in culture sustain viability and function through preservation of oxidant status [41], and preconditioning with kallikrein [26] or melatonin [27] enhances their therapeutic potential.

An important challenge for clinical translation is the risk for long-term MSC maldifferentiation. While intrarenal injection of rat MSCs initially preserves renal function in a rat model of glomerulonephritis, a significant proportion of the glomeruli subsequently contained large adipocytes with glomerular sclerosis [42]. Furthermore, reports of sarcoma [43] and teratoma [44] arising from exogenous MSCs illustrate their potential for transformation into tumors, underscoring the requirement for closely monitoring human MSCs in clinical studies. Alternatively, complications and maldifferentiation of live replicating MSCs warrant development of safer tactics and interventions.

Considerable evidence shows that MSCs release microvesicles which exhibit characteristics of their parental cells, and transfer proteins, lipids, and genetic material to target cells. We have recently shown that endothelial outgrowth cells release microvesicles [

], which may mediate their intercellular communications. Similarly, MSCs are avid producers of microvesicles [

] (Figure

) that shuttle functional components for their paracrine action [

]. Delivery of microvesicles instead of their parent MSCs could avoid concerns about extensive expansion, cryopreservation, complications, and maldifferentiation of live replicating cells. Indeed, microvesicles derived from preconditioned MSCs promoted recovery in a rat hind-limb ischemia model [

]. However, questions regarding their composition and potency relative to their parent MSCs remain unanswered, underscoring the need for studies to clarify the potential of this promising therapeutic modality.

Mesenchymal stem cell release microvesicles. Transmission electron microscopy image (left) and scanning electron microscopy image (right) showing release of microvesicles (arrows) from adipose tissue-derived mesenchymal stem cells (26,500).

Uremic conditions may also affect the efficacy of MSCs, limiting their potential use in patients with CKD. Uremia induced by partial kidney ablation in C57Bl/6J mice leads to MSC functional incompetence, characterized by decreased expression of VEGF, VEGF receptor-1, and stromal derived factor-1, increased cellular senescence, and decreased proliferation [49]. Conversely, MSCs isolated from subcutaneous adipose tissue of healthy controls and patients with renal disease show similar characteristics and functionality, underscoring the feasibility of autologous cell therapy in patients with renal disease [50]. Indeed, a recent meta-analysis of prospective clinical trials that used intravascular delivery of MSCs concluded that these cells have an excellent safety record [51].

Although it is accepted that MSCs from different species are capable of differentiation into various lineages and express common MSC markers, species-dependent variability in their expression has been reported among different species [52]. Furthermore, the mechanism of MSC-mediated immunosuppression varies among different species. For example, while immunosuppression by human-derived or monkey-derived MSCs is mediated by indoleamine 2,3-dioxygenase, the molecular mechanisms underlying immunosuppression in mouse MSCs utilize nitric oxide [53]. Several immune barriers have been also encountered in experimental xenotransplantation, the transplantation of MSCs from one species to another, warranting the development of genetic alternatives to overcome these obstacles [54]. Clearly, results from experimental studies need to be carefully validated before clinical translation.

There is also a pressing need for better methods for detection and monitoring the fate of MSCs. Despite improvement in direct (fluorescent probe) [55] and indirect (reporter genes) [56] labeling techniques, questions regarding interactions of MSCs with tissue, differentiation, or migration remain unanswered. While fluorescent probes such as membrane tracers or microspheres need to be detected with histological techniques in a cell or organelle, reporter genes such as bioluminescence or fluorescent proteins can be used to identify different cell populations using imaging in vivo[57, 58]. However, these detection methods have little tissue penetration, limiting their use in large animal models or humans [59].

Conceivably, imaging modalities such as single-photon emission computed tomography or magnetic resonance imaging may address some of these deficiencies by providing high-resolution anatomical detail and tracking of cell viability [60, 61]. Several types of agents are currently used for labeling MSCs for their detection with magnetic resonance imaging. Among them, superparamagnetic iron oxide particles are the most commonly applied, because of their capacity to induce changes in T2 relaxivity in vivo[62]. However, the transfection agents used for superparamagnetic iron oxide particle internalization may also affect cell viability, and dying cells accumulate iron until dissolved or eliminated by phagocytosis, impeding their application as indices of cell viability. Further methods are therefore needed to better assess engraftment, survival, and function of MSCs in human subjects.

More:
Mesenchymal stem cell treatment for chronic renal failure ...

Read More...

Page 7«..6789..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick