Home | Sources Directory | News Releases | Calendar | Articles | | Contact |  

Stem cell treatments

Stem cell treatments are a type of intervention strategy that introduces new cells into damaged tissue in order to treat disease or injury. Many medical researchers believe that stem cell treatments have the potential to change the face of human disease and alleviate suffering.[citation needed] The ability of stem cells to self-renew and give rise to subsequent generations with variable degrees of differentiation capacities, [1] offers significant potential for generation of tissues that can potentially replace diseased and damaged areas in the body, with minimal risk of rejection and side effects.

A number of stem cell therapeutics exist, but most are at experimental stages and/or costly, with the notable exception of bone marrow transplantation.[citation needed] Medical researchers anticipate that adult and embryonic stem cells will soon be able to treat cancer, Type 1 diabetes mellitus, Parkinson's disease, Huntington's disease,Celiac Disease, cardiac failure, muscle damage and neurological disorders, and many others.[2] Nevertheless, before stem cell therapeutics can be applied in the clinical setting, more research is necessary to understand stem cell behavior upon transplantation as well as the mechanisms of stem cell interaction with the diseased/injured microenvironment.[2]

Contents

[edit] Current treatments

For over 30 years, bone marrow, and more recently, umbilical cord blood stem cells, have been used to treat cancer patients with conditions such as leukemia and lymphoma.[3] During chemotherapy, most growing cells are killed by the cytotoxic agents. These agents, however, cannot discriminate between the leukemia or neoplastic cells, and the hematopoietic stem cells within the bone marrow. It is this side effect of conventional chemotherapy strategies that the stem cell transplant attempts to reverse; a donor's healthy bone marrow reintroduces functional stem cells to replace the cells lost in the host's body during treatment.

[edit] Potential treatments

[edit] Brain damage

Stroke and traumatic brain injury lead to cell death, characterized by a loss of neurons and oligodendrocytes within the brain. Healthy adult brains contain neural stem cells which divide to maintain general stem cell numbers, or become progenitor cells. In healthy adult animals, progenitor cells migrate within the brain and function primarily to maintain neuron populations for olfaction (the sense of smell). Interestingly, in pregnancy and after injury, this system appears to be regulated by growth factors and can increase the rate at which new brain matter is formed.[citation needed] Although the reparative process appears to initiate following trauma to the brain, substantial recovery is rarely observed in adults, suggesting a lack of robustness.

Stem cells may also be used to treat brain degeneration, such as in Parkinson's and Alzheimer's disease.[4][5]

[edit] Cancer

Research injecting neural (adult) stem cells into the brains of dogs has shown to be very successful in treating cancerous tumors.[citation needed] Using conventional techniques, brain cancer is difficult to treat because it spreads so rapidly. Researchers at the Harvard Medical School transplanted human neural stem cells into the brain of rodents that received intracranial tumours. Within days, the cells migrated into the cancerous area and produced cytosine deaminase, an enzyme that converts a non-toxic pro-drug into a chemotheraputic agent. As a result, the injected substance was able to reduce the tumor mass by 81 percent. The stem cells neither differentiated nor turned tumorigenic.[6] Some researchers believe that the key to finding a cure for cancer is to inhibit proliferation of cancer stem cells. Accordingly, current cancer treatments are designed to kill cancer cells. However, conventional chemotherapy treatments cannot discriminate between cancerous cells and others. Stem cell therapies may serve as potential treatments for cancer.[7]

[edit] Spinal cord injury

A team of Korean researchers reported on November 25, 2003, that they had transplanted multipotent adult stem cells from umbilical cord blood to a patient suffering from a spinal cord injury and that following the procedure, she could walk on her own, without difficulty. The patient had not been able to stand up for roughly 19 years. For the unprecedented clinical test, the scientists isolated adult stem cells from umbilical cord blood and then injected them into the damaged part of the spinal cord.[8] [9]

According to the October 7, 2005 issue of The Week, University of California, Irvine researchers transplanted multipotent human fetal-derived neural stem cells into paralyzed mice, resulting in locomotor improvements four months later. The observed recovery was associated with differentiation of transplanted cells into new neurons and oligodendrocytes- the latter of which forms the myelin sheath around axons of the central nervous system, thus insulating neural impulses and facilitating communication with the brain.[10]

In January 2005, researchers at the University of Wisconsin'Madison differentiated human blastocyst stem cells into neural stem cells, then into pre-mature motor neurons, and finally into spinal motor neurons, the cell type that, in the human body, transmits messages from the brain to the spinal cord and subsequently mediates motor function in the periphery. The newly generated motor neurons exhibited electrical activity, the signature action of neurons. Lead researcher Su-Chun Zhang described the process as "[teaching] the blastocyst stem cells to change step by step, where each step has different conditions and a strict window of time."

Transformation of blastocyst stem cells into motor neurons had eluded researchers for decades. While Zhang's findings were a significant contribution to the field, the ability of transplanted neural cells to establish communication with neighboring cells remains unclear. Accordingly, studies using chicken embryos as a model organism can be an effective proof-of-concept experiment. If functional, the new cells could be used to treat diseases like Lou Gehrig's disease, muscular dystrophy, and spinal cord injuries.[citation needed]

[edit] Heart damage

Several clinical trials targeting heart disease have shown that adult stem cell therapy is safe, effective, and equally efficient in treating old and recent infarcts.[11] Adult stem cell therapy for treating heart disease was commercially available in at least five continents at the last count (2007).

Possible mechanisms of recovery include:[4]

  • Generation of heart muscle cells
  • Stimulation of growth of new blood vessels to repopulate damaged heart tissue
  • Secretion of growth factors
  • Assistance via some other mechanism

It may be possible to have adult bone marrow cells differentiate into heart muscle cells.[4]

[edit] Haematopoiesis (blood cell formation)

The specificity of the human immune cell repertoire is what allows the human body to defend itself from rapidly adapting antigens. However, the immune system is vulnerable to degradation upon the pathogenesis of disease, and because of the critical role that it plays in overall defense, its degradation is often fatal to the organism as a whole. Diseases of hematopoietic cells are called hematopathology. The specificity of the immune cells is what allows recognition of foreign antigens, causing further challenges in the treatment of immune disease. Identical matches between donor and recipient must be made for successful transplantation treatments, but matches are uncommon, even between first-degree relatives. Research using both hematopoietic adult stem cells and embryonic stem cells has provided insight into the possible mechanisms and methods of treatment for many of these ailments.[citation needed]

Fully mature human red blood cells may be generated ex vivo by hematopoietic stem cells (HSCs), which are precursors of red blood cells. In this process, HSCs are grown together with stromal cells, creating an environment that mimics the conditions of bone marrow, the natural site of red blood cell growth. Erythropoietin, a growth factor, is added, coaxing the stem cells to complete terminal differentiation into red blood cells.[12] Further research into this technique should have potential benefits to gene therapy, blood transfusion, and topical medicine.

[edit] Baldness

Hair follicles also contain stem cells, and some researchers predict research on these follicle stem cells may lead to successes in treating baldness through "hair multiplication", also known as "hair cloning". This treatment is expected to work by taking stem cells from existing follicles, multiplying them in culture, and implanting the new follicles back into the scalp. Later treatments may be able to simply signal follicle stem cells to give off chemical signals to nearby follicle cells which have shrunk during the aging process, which in turn respond to these signals by regenerating and once again making healthy hair.[13]

[edit] Missing teeth

In 2004, scientists at King's College London discovered a way to cultivate a complete tooth in mice[14] and were able to grow them stand-alone in the laboratory. Researchers are confident that this technology can be used to grow live teeth in human patients.

In theory, stem cells taken from the patient could be coaxed in the lab into turning into a tooth bud which, when implanted in the gums, will give rise to a new tooth, and would be expected to grow within two months.[15] It will fuse with the jawbone and release chemicals that encourage nerves and blood vessels to connect with it. The process is similar to what happens when humans grow their original adult teeth.

Many challenges remain, however, before stem cells could be a choice for the replacement of missing teeth in the future.[16]

[edit] Deafness

Heller has reported success in re-growing cochlea hair cells with the use of embryonic stem cells.[17]

[edit] Blindness and vision impairment

Since 2003, researchers have successfully transplanted corneal stem cells into damaged eyes to restore vision. Using embryonic stem cells, scientists are able to grow a thin sheet of totipotent stem cells in the laboratory. When these sheets are transplanted over the damaged cornea, the stem cells stimulate renewed repair, eventually restore vision.[18] The latest such development was in June 2005, when researchers at the Queen Victoria Hospital of Sussex, England were able to restore the sight of forty patients using the same technique. The group, led by Dr. Sheraz Daya, was able to successfully use adult stem cells obtained from the patient, a relative, or even a cadaver. Further rounds of trials are ongoing.[19]

In April 2005, doctors in the UK transplanted corneal stem cells from an organ donor to the cornea of Deborah Catlyn, a woman who was blinded in one eye when acid was thrown in her eye at a nightclub. The cornea, which is the transparent window of the eye, is a particularly suitable site for transplants. In fact, the first successful human transplant was a cornea transplant. The absence of blood vessels within the cornea makes this area a relatively easy target for transplantation. The majority of corneal transplants carried out today are due to a degenerative disease called keratoconus.

The University Hospital of New Jersey reports that the success rate for growth of new cells from transplanted stem cells varies from 25 percent to 70 percent.[20]

In 2009, researchers at the University of Pittsburgh Medical center demonstrated that stem cells collected from human corneas can restore transparency without provoking a rejection response in mice with corneal damage. [21]

[edit] Amyotrophic lateral sclerosis

Stem cells have resulted in significant locomotor improvements in rats with an Amyotrophic lateral sclerosis-like disease. In a rodent model that closely mimics the human form of ALS, animals were injected with a virus to kill the spinal cord motor nerves which mediate movement. Animals subsequently received stem cells in the spinal cord. Transplanted cells migrated to the sites of injury, contributed to regeneration of the ablated nerve cells, and restored locomotor function.[22]

[edit] Graft vs. host disease and Crohn's disease

Phase III clinical trials expected to end in second-quarter 2008 were conducted by Osiris Therapeutics using their in-development product Prochymal, derived from adult bone marrow. The target disorders of this therapeutic are graft-versus-host disease and Crohn's disease.[23]

[edit] Neural and behavioral birth defects

A team of researchers led by Prof. Joseph Yanai were able to reverse learning deficits in the offspring of pregnant mice who were exposed to heroin and the pesticide organophosphate.[citation needed] This was done by direct neural stem cell transplantation into the brains of the offspring. The recovery was almost 100 percent, as shown in behavioral tests that suggested improved to normal behavior and learning scores in animals receiving cell transplantation.[clarification needed] On the molecular level, brain chemistry of the treated animals was also restored to normal. Through the work, which was supported by the US National Institutes of Health, the US-Israel Binational Science Foundation and the Israel anti-drug authorities, the researchers discovered that the stem cells worked even in cases where most of the cells died out in the host brain.

The scientists found that before they die the neural stem cells succeed in inducing the host brain to produce large numbers of stem cells which repair the damage. These findings, which answered a major question in the stem cell research community, were published earlier this year in the leading journal, Molecular Psychiatry. Scientists are now developing procedures to administer the neural stem cells in the least invasive way possible - probably via blood vessels, making therapy practical and clinically feasible. Researchers also plan to work on developing methods to take cells from the patient's own body, turn them into stem cells, and then transplant them back into the patient's blood via the blood stream. Aside from decreasing the chances of immunological rejection, the approach will also eliminate the controversial ethical issues involved in the use of stem cells from human embryos.[24]

[edit] Diabetes

Diabetes patients lose the function of insulin-producing beta cells within the pancreas. Human embryonic stem cells may be grown in cell culture and stimulated to form insulin-producing cells that can be transplanted into the patient.

However, clinical success is highly dependent on the development of the following procedures:[4]

  • Transplanted cells should proliferate
  • Transplanted cells should differentiate in a site-specific manner
  • Transplanted cells should survive in the recipient (prevention of transplant rejection)
  • Transplanted cells should integrate within the targeted tissue
  • Transplanted cells should integrate into the host circuitry and restore function

[edit] Orthopaedics

Clinical case reports in the treatment of orthopaedic conditions have been reported. To date, the focus in the literature for musculoskeletal care appears to be on mesenchymal stem cells. Centeno et al. have published MRI evidence of increased cartilage and meniscus volume in individual human subjects.[25] [26] The results of trials that include a large number of subjects, are yet to be published. However, a published safety study conducted in a group of 227 patients over a 3-4 year period shows adequate safety and minimal complications associated with mesenchymal cell transplantation.[27]

Wakitani has also published a small case series of nine defects in five knees involving surgical transplantation of mesenchymal stem cells with coverage of the treated chondral defects.[28]

[edit] Wound healing

Stem cells can also be used to stimulate the growth of human tissues. In an adult, wounded tissue is most often replaced by scar tissue, which is characterized in the skin by disorganized collagen structure, loss of hair follicles and irregular vascular structure. In the case of wounded fetal tissue, however, wounded tissue is replaced with normal tissue through the activity of stem cells.[29] A possible method for tissue regeneration in adults is to place adult stem cell "seeds" inside a tissue bed "soil" in a wound bed and allow the stem cells to stimulate differentiation in the tissue bed cells. This method elicits a regenerative response more similar to fetal wound-healing than adult scar tissue formation.[29] Researchers are still investigating different aspects of the "soil" tissue that are conducive to regeneration.[29]

[edit] Infertility

Culture of human embryonic stem cells in mitotically inactivated porcine ovarian fibroblasts (POF) causes differentiation into germ cells (precursor cells of oocytes and spermatozoa), as evidenced by gene expression analysis.[30]

Human embryonic stem cells have been stimulated to form Spermatozoon-like cells, yet still slightly damaged or malformed.[31] It could potentially treat azoospermia.

[edit] Clinical Trials

On January 23, 2009, the US Food and Drug Administration gave clearance to Geron Corporation for the initiation of the first clinical trial of an embryonic stem cell-based therapy on humans. The trial will evaluate the drug GRNOPC1, embryonic stem cell-derived oligodendrocyte progenitor cells, on patients with acute spinal cord injury.[32]

As of mid 2010 hundreds of phase III clinical trials involving stem cells have been registered.[33]

[edit] Stem cell use in animals

[edit] Veterinary applications

[edit] Potential contributions to veterinary medicine

Research currently conducted on horses, dogs, and cats can benefit the development of stem-cell treatments in veterinary medicine and can target a wide range of injuries and diseases such as myocardial infarction, stroke, tendon and ligament damage, osteoarthritis, osteochondrosis and muscular dystrophy both in large animals, as well as humans.[34][35][36][37] While investigation of cell-based therapeutics generally reflects human medical needs, the high degree of frequency and severity of certain injuries in racehorses has put veterinary medicine at the forefront of this novel regenerative approach.[38] Companion animals can serve as clinically relevant models that closely mimic human disease.[39][40]

[edit] Development of regenerative treatment models

Veterinary applications of stem cell therapy as a means of tissue regeneration have been largely shaped by research that began with the use of adult-derived mesenchymal stem cells to treat animals with injuries or defects affecting bone, cartilage, ligaments and/or tendons.[41][42][43] Because mesenchymal stem cells can differentiate into the cells that make up bone, cartilage, tendons, and ligaments (as well as muscle, fat, and possibly other tissues), they have been the main type of stem cells studied in the treatment of diseases affecting these tissues.[44][42] Mesenchymal stem cells are primarily derived from adipose tissue or bone marrow. Since an elevated immune response following cell transplantation may result in rejection of exogenous cells (except in the case of cells derived from a very closely genetically related individual), mesenchymal stem cells are often derived from the patient prior to injection in a process known as autologous transplantation.[45] Surgical repair of bone fractures in dogs and sheep has demonstrated that engraftment of mesenchymal stem cells derived from a genetically different donor within the same species, termed allogeneic transplantation, does not elicit an immunological response in the recipient animal and can mediate regeneration of bone tissue in major bony fractures and defects.[46][47] Stem cells can speed up bone repair in fractures/defects that would normally require extensive grafting, suggesting that mesenchymal stem cell use may provide a useful alternative to conventional grafting techniques.[46][47] Treating tendon and ligament injuries in horses using stem cells, whether derived from adipose tissue or bone-marrow, has support in the veterinary literature.[48][49] While further studies are necessary to fully characterize the use of cell-based therapeutics for treatment of bone fractures, stem cells are thought to mediate repair via five primary mechanisms: 1) providing an antiinflammatory effect, 2) homing to damaged tissues and recruiting other cells, such as endothelial progenitor cells, that are necessary for tissue growth, 3) supporting tissue remodeling over scar formation, 4) inhibiting apoptosis, and 5) differentiating into bone, cartilage, tendon, and ligament tissue.[49][50]
[edit] Significance of stem cell microenvironments
The microenvironment into which stem cells are transplanted significantly alters the capacity of engrafted cells for recovery and repair. The microenviroment provides growth factors and other chemical signals that guide appropriate differentiation of transplanted cell populations and direct transplanted cells to sites of trauma or disease. Repair and recovery can then be mediated via three primary mechanisms: 1) formation and/or recruitment of new blood cells to the damaged region; 2) prevention of programed cell death or apoptosis; and 3) suppression of inflammation.[45][47][2] To further enrich blood supply to the damaged areas, and consequently promote tissue regeneration, platelet-rich plasma could be used in conjunction with stem cell transplantation.[45][51] The efficacy of some stem cell populations may also be affected by the method of delivery; for instance, to regenerate bone, stem cells are often introduced in a scaffold where they produce the minerals necessary for generation of functional bone.[45][47][2][51]
[edit] Sources of autologous (patient-derived) stem cells
Autologous stem cells intended for regenerative therapy are generally isolated either from the patient's bone marrow or from adipose tissue. The number of stem cells transplanted into damaged tissue may alter efficacy of treatment. Accordingly, stem cells derived from bone marrow aspirates, for instance, are cultured in specialized laboratories for expansion to millions of cells.[47][51] Although adipose-derived tissue also requires processing prior to use, the culturing methodology for adipose-derived stem cells is not as extensive as that for bone marrow-derived cells.[52][53] While it is thought that bone-marrow derived stem cells are preferred for bone, cartilage, ligament, and tendon repair, others believe that the less challenging collection techniques and the multi-cellular microenvironment already present in adipose-derived stem cell fractions make the latter the preferred source for autologous transplantation.[45]

[edit] Currently Available Treatments for Horses and Dogs Suffering from Orthopedic Conditions

Autologous or allogeneic stem cells are currently used as an adjunctive therapy in the surgical repair of some types of fractures in dogs and horses.[47][54] Autologous stem cell-based treatments for ligament injury, tendon injury, osteoarthritis, osteochondrosis, and sub-chondral bone cysts have been commercially available to practicing veterinarians to treat horses since 2003 in the United States and since 2006 in the United Kingdom. Autologous stem-cell based treatments for tendon injury, ligament injury, and osteoarthritis in dogs have been available to veterinarians in the United States since 2005. Over 3000 privately-owned horses and dogs have been treated with autologous adipose-derived stem cells. The efficacy of these treatments has been shown in double-blind clinical trials for dogs with osteoarthritis of the hip and elbow and horses with tendon damage.[55][56] The efficacy of using stem cells, whether adipose-derived or bone-marrow derived, for treating tendon and ligament injuries in horses has support in the veterinary literature.[48][49]

[edit] Developments in Stem Cell Treatments in Veterinary Internal Medicine

Currently, research is being conducted to develop stem cell treatments for: 1) horses suffering from COPD, neurologic disease, and laminitis; and 2) dogs and cats suffering from heart disease, liver disease, kidney disease, neurologic disease, and immune-mediated disorders.

[edit] Embryonic stem cell controversy

There is wide-spread controversy over the use of human embryonic stem cells. This controversy primarily targets the techniques used to derive new embryonic stem cell lines, which often requires the destruction of the blastocyst.

Opposition to the use of human embryonic stem cells in research is often based on philosophical, moral or religious objections. At present, there are alternative sources for stem cells which have achieved considerable success when used as medical therapies. These alternatives do not require the destruction of an embryo, such as the use of umbilical cord blood, milky teeth stem cells, bone marrow stem cells or using induced pluripotent stem cells. However, non-embryonic stem cells may have limitations their embryonic counterparts do not.

[edit] Stem cell treatments around the world

[edit] China

Stem cell research and treatment is currently being practiced at a clinical level in the People's Republic of China. The Ministry of Health of the People's Republic of China has permitted the use of stem cell therapy for conditions beyond those approved of in Western countries such as the United States, United Kingdom, and Australia. However, stem cell therapy in China has received scrutiny for its failed attempts to meet international standards of safety and efficacy. [57]

Stem cell therapies provided in China utilize a variety of cell types including umbilical cord stem cells and olfactory ensheathing cells. The stem cells are then expanded in centralized blood banks before being used in stem cell treatments. State-funded companies based in the Shenzhen Hi-Tech Industrial Zone claim to treat the symptoms of numerous disorders with adult stem cell therapy. Hospitals throughout eastern China provide numerous therapies to patients in coordination with the stem cell providers. These companies' therapies are currently focused on the treatment of neurodegenerative and cardiovascular disorders. However, retrospective studies have shown that Chinese use of fetal-derived brain tissue in spinal cord injured human subjects were not as promising as once thought: the phenotype and the fate of the transplanted cells, described as olfactory ensheathing cells, were unknown. As well, perioperative morbidity and lack of functional benefit were identified as the most serious clinical shortcomings.[58] Furthermore, the extent of regulatory policy in the use of stem cell therapies in China is unclear.[59] Thus, in the absence of a valid clinical trials protocol, and more regulatory oversight, patients and physicians are advised to be highly cautious when selecting Chinese stem cell therapeutics.[60]

[edit] Mexico

Stem cell treatment is currently being practiced at a clinical level in Mexico. An International Health Department Permit (COFEPRIS) is required. This permit allows the use of stem cell types beyond those approved of in Western countries such as the United States or Europe. Stem cell therapies provided in Mexico utilize patient Adipose, Bone Marrow, or Donor Placenta sources.[61]

[edit] South Korea

In 2005, South Korean scientists claimed to have generated stem cells that were tailored to match the recipient. Each of the 11 new stem cell lines was developed using somatic cell nuclear transfer (SCNT) technology. The resultant cells were thought to match the genetic material of the recipient, thus suggesting minimal to no cell rejection.[62]

This study, however, was eventually discredited as the primary researcher,Dr. Woo Suk Hwang, admitted to using cells obtained from his research staff.[citation needed] In Dec 2005, claims were put forward that his research had been manipulated to wrongfully indicate positive results. Later that month, these claims were confirmed by an academic panel.[63]

[edit] See also

[edit] External links

[edit] References

  1. ^ Weissman IL (January 2000). "Stem cells: units of development, units of regeneration, and units in evolution". Cell 100 (1): 157'68. doi:10.1016/S0092-8674(00)81692-X. PMID 10647940. http://linkinghub.elsevier.com/retrieve/pii/S0092-8674(00)81692-X.  as cited in Gurtner GC, Callaghan MJ, Longaker MT (2007). "Progress and potential for regenerative medicine". Annu. Rev. Med. 58: 299'312. doi:10.1146/annurev.med.58.082405.095329. PMID 17076602. http://arjournals.annualreviews.org/doi/abs/10.1146/annurev.med.58.082405.095329?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%3dncbi.nlm.nih.gov. 
  2. ^ a b c d Singec I, Jandial R, Crain A, Nikkhah G, Snyder EY (2007). "The leading edge of stem cell therapeutics". Annu. Rev. Med. 58: 313'28. doi:10.1146/annurev.med.58.070605.115252. PMID 17100553. http://arjournals.annualreviews.org/doi/abs/10.1146/annurev.med.58.070605.115252?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%3dncbi.nlm.nih.gov. 
  3. ^ Bone Marrow Transplantation and Peripheral Blood Stem Cell Transplantation In National Cancer Institute Fact Sheet web site. Bethesda, MD: National Institutes of Health, U.S. Department of Health and Human Services, 2010. Cited August 24, 2010
  4. ^ a b c d Cell Basics: What are the potential uses of human stem cells and the obstacles that must be overcome before these potential uses will be realized?. In Stem Cell Information World Wide Web site. Bethesda, MD: National Institutes of Health, U.S. Department of Health and Human Services, 2009. cited Sunday, April 26, 2009
  5. ^ http://www.sciencedaily.com/releases/2009/07/090720190726.htm
  6. ^ Stem Cells Tapped to Replenish Organs thescientist.com, Nov 2000. By Douglas Steinberg
  7. ^ "Cancer Stem Cells Hint at Cure" at wired.com
  8. ^ Kang KS, Kim SW, Oh YH, et al. (2005). "A 37-year-old spinal cord-injured female patient, transplanted of multipotent stem cells from human UC blood, with improved sensory perception and mobility, both functionally and morphologically: a case study". Cytotherapy 7 (4): 368'73. doi:10.1080/14653240500238160. PMID 16162459. 
  9. ^ team co-headed by researchers at Chosun University, Seoul National University and the Seoul Cord Blood Bank (SCB) Umbilical cord cells 'allow paralysed woman to walk' By Roger Highfield, Science Editor. Last Updated: 1:28AM GMT 30 Nov 2004
  10. ^ http://www.ncbi.nlm.nih.gov/pubmed/16172374
  11. ^ Strauer BE, Schannwell CM, Brehm M (April 2009). "Therapeutic potentials of stem cells in cardiac diseases". Minerva Cardioangiol 57 (2): 249'67. PMID 19274033. 
  12. ^ Giarratana MC, Kobari L, Lapillonne H, et al. (January 2005). "Ex vivo generation of fully mature human red blood cells from hematopoietic stem cells". Nat. Biotechnol. 23 (1): 69'74. doi:10.1038/nbt1047. PMID 15619619. 
  13. ^ Hair Cloning Nears Reality as Baldness Cure WebMD November 2004
  14. ^ The Daily Telegraph (London). http://www.telegraph.co.uk/connected/main.jhtml?view=DETAILS&grid=P8&targetRule=10&xml=%2Fconnected%2F2004%2F02%2F17%2Fecntee15.xml. Retrieved May 24, 2010. 
  15. ^ Teeth from scratch
  16. ^ Yen AH, Sharpe PT (January 2008). "Stem cells and tooth tissue engineering". Cell Tissue Res. 331 (1): 359'72. doi:10.1007/s00441-007-0467-6. PMID 17938970. 
  17. ^ Gene therapy is first deafness 'cure' - health - 14 February 2005 - New Scientist
  18. ^ Fetal tissue restores lost sight MedicalNewsToday. Article Date: 28 Oct 2004 - 10:00 PDT
  19. ^ BBC NEWS | England | Southern Counties | Stem cells used to restore vision
  20. ^ [1] The University Hospital of New Jersey, 2002
  21. ^ "Stem Cell Therapy Makes Cloudy Corneas Clear, According To Pitt Researchers". Medical News Today. 13 April 2009. http://www.medicalnewstoday.com/articles/145528.php. Retrieved 2009-06-04. 
  22. ^ Vastag B (April 2001). "Stem cells step closer to the clinic: paralysis partially reversed in rats with ALS-like disease". JAMA 285 (13): 1691'3. doi:10.1001/jama.285.13.1691. PMID 11277806. http://jama.ama-assn.org/cgi/pmidlookup?view=long&pmid=11277806. 
  23. ^ Querida Anderson (2008-06-15). "Osiris Trumpets Its Adult Stem Cell Product". Genetic Engineering & Biotechnology News (Mary Ann Liebert, Inc.): p. 13. http://www.genengnews.com/articles/chitem.aspx?aid=2508. Retrieved 2008-07-06. "(subtitle) Procymal is being developed in many indications, GvHD being the most advanced" 
  24. ^ [2] ISRAEL21c > Israeli scientists reverse brain birth defects using stem cells. December 25, 2008. (Researchers from the Hebrew University of Jerusalem-Hadassah Medical led by Prof. Joseph Yanai)
  25. ^ Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman M, Karli D (December 2008). "Regeneration of meniscus cartilage in a knee treated with percutaneously implanted autologous mesenchymal stem cells". Med. Hypotheses 71 (6): 900'8. doi:10.1016/j.mehy.2008.06.042. PMID 18786777. 
  26. ^ Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman M, Karli D (2008). "Increased knee cartilage volume in degenerative joint disease using percutaneously implanted, autologous mesenchymal stem cells". Pain Physician 11 (3): 343'53. PMID 18523506. http://www.painphysicianjournal.com/linkout_vw.php?issn=1533-3159&vol=11&page=343. 
  27. ^ Centeno CJ, Schultz JR, Cheever M, Robinson B, Freeman M, Marasco W (March 2010). "Safety and complications reporting on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique". Curr Stem Cell Res Ther 5 (1): 81'93. doi:10.2174/157488810790442796. PMID 19951252. http://www.bentham-direct.org/pages/content.php?CSCR/2010/00000005/00000001/0011CSCR.SGM. 
  28. ^ Wakitani S, Nawata M, Tensho K, Okabe T, Machida H, Ohgushi H (2007). "Repair of articular cartilage defects in the patello-femoral joint with autologous bone marrow mesenchymal cell transplantation: three case reports involving nine defects in five knees". J Tissue Eng Regen Med 1 (1): 74'9. doi:10.1002/term.8. PMID 18038395. 
  29. ^ a b c Gurtner GC, Callaghan, MJ and Longaker MT. 2007. Progress and potential for regenerative medicine. Annu. Rev. Med 58:299-312
  30. ^ Richards M, Fong CY, Bongso A (December 2008). "Comparative evaluation of different in vitro systems that stimulate germ cell differentiation in human embryonic stem cells". Fertil. Steril. 93 (3): 986'94. doi:10.1016/j.fertnstert.2008.10.030. PMID 19064262. 
  31. ^ Ledford H (7 July 2009). "Sperm-like cells made from human embryonic stem cells" ([dead link]). Nature News. doi:10.1038/news.2009.646. http://www.nature.com/news/2009/070709/full/news.2009.646.html. 
  32. ^ http://www.geron.com/products/productinformation/spinalcordinjury.aspx
  33. ^ http://www.clinicaltrials.gov/ct2/results?term=stem+cell&phase=2
  34. ^ Chen J, Li Y, Wang L, et al. (April 2001). "Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats". Stroke 32 (4): 1005'11. PMID 11283404. http://stroke.ahajournals.org/cgi/pmidlookup?view=long&pmid=11283404. 
  35. ^ Assmus B, Schächinger V, Teupe C, et al. (December 2002). "Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI)". Circulation 106 (24): 3009'17. doi:10.1161/01.CIR.0000043246.74879.CD. PMID 12473544. http://circ.ahajournals.org/cgi/pmidlookup?view=long&pmid=12473544. 
  36. ^ Murphy JM, Fink DJ, Hunziker EB, Barry FP (December 2003). "Stem cell therapy in a caprine model of osteoarthritis". Arthritis Rheum. 48 (12): 3464'74. doi:10.1002/art.1136510.1002/art.11365. PMID 14673997. 
  37. ^ Sampaolesi M, Blot S, D'Antona G, et al. (November 2006). "Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs". Nature 444 (7119): 574'9. doi:10.1038/nature0528210.1038/nature05282. PMID 17108972. 
  38. ^ Taylor SE, Smith RK, Clegg PD (March 2007). "Mesenchymal stem cell therapy in equine musculoskeletal disease: scientific fact or clinical fiction?". Equine Vet. J. 39 (2): 172'80. doi:10.2746/042516407X180868. PMID 17378447. 
  39. ^ Tecirlioglu RT, Trounson AO (2007). "Embryonic stem cells in companion animals (horses, dogs and cats): present status and future prospects". Reprod. Fertil. Dev. 19 (6): 740'7. doi:10.1071/RD07039. PMID 17714628. http://www.publish.csiro.au/journals/abstractHTML.cfm?J=RD&V=19&I=6&F=RD07039abs.XML. 
  40. ^ Koch TG, Betts DH (November 2007). "Stem cell therapy for joint problems using the horse as a clinically relevant animal model". Expert Opin Biol Ther 7 (11): 1621'6. doi:10.1517/14712598.7.11.1621. PMID 17961087. http://informahealthcare.com/doi/abs/10.1517/14712598.7.11.1621%20. 
  41. ^ Young RG, Butler DL, Weber W, Caplan AI, Gordon SL, Fink DJ (July 1998). "Use of mesenchymal stem cells in a collagen matrix for Achilles tendon repair". J. Orthop. Res. 16 (4): 406'13. doi:10.1002/jor.1100160403. PMID 9747780. 
  42. ^ a b Awad HA, Butler DL, Boivin GP, et al. (June 1999). "Autologous mesenchymal stem cell-mediated repair of tendon". Tissue Eng. 5 (3): 267'77. doi:10.1089/ten.1999.5.267. PMID 10434073. 
  43. ^ Bruder SP, Kraus KH, Goldberg VM, Kadiyala S (July 1998). "The effect of implants loaded with autologous mesenchymal stem cells on the healing of canine segmental bone defects". J Bone Joint Surg Am 80 (7): 985'96. PMID 9698003. http://www.ejbjs.org/cgi/pmidlookup?view=long&pmid=9698003. 
  44. ^ Nathan S, Das De S, Thambyah A, Fen C, Goh J, Lee EH (August 2003). "Cell-based therapy in the repair of osteochondral defects: a novel use for adipose tissue". Tissue Eng. 9 (4): 733'44. doi:10.1089/10763270376824741210.1089/107632703768247412. PMID 13678450. 
  45. ^ a b c d e Kane, Ed (May 2008). Stem cell therapy shows promise for soft-tissue injury, disease. DVM Newsmagazine. 6E-10E.
  46. ^ a b Kraus KH, Kirker-Head C (April 2006). "Mesenchymal stem cells and bone regeneration". Vet Surg 35 (3): 232'42. doi:10.1111/j.1532-950X.2006.00142.x. PMID 16635002. http://www3.interscience.wiley.com/resolve/openurl?genre=article&sid=nlm:pubmed&issn=0161-3499&date=2006&volume=35&issue=3&spage=232. 
  47. ^ a b c d e f Zachos TA, Smith TJ (September 2008). Use of adult stem cells in clinical orthopedics. DVM Newsmagazine. 36-39.
  48. ^ a b Smith RKW (2008). "Principles of stem cell therapy in the horse ' the science behind the technology". Pferdeheilkunde 24 (4): 508. 
  49. ^ a b c Richardson LE, Dudhia J, Clegg PD, Smith R (September 2007). "Stem cells in veterinary medicine'attempts at regenerating equine tendon after injury". Trends Biotechnol. 25 (9): 409'16. doi:10.1016/j.tibtech.2007.07.009. PMID 17692415. http://linkinghub.elsevier.com/retrieve/pii/S0167-7799(07)00188-6. 
  50. ^ Csaki C, Matis U, Mobasheri A, Ye H, Shakibaei M (December 2007). "Chondrogenesis, osteogenesis and adipogenesis of canine mesenchymal stem cells: a biochemical, morphological and ultrastructural study". Histochem. Cell Biol. 128 (6): 507'20. doi:10.1007/s00418-007-0337-z10.1007/s00418-007-0337-z. PMID 17922135. 
  51. ^ a b c Yamada Y, Ueda M, Naiki T, Takahashi M, Hata K, Nagasaka T (2004). "Autogenous injectable bone for regeneration with mesenchymal stem cells and platelet-rich plasma: tissue-engineered bone regeneration". Tissue Eng. 10 (5-6): 955'64. doi:10.1089/107632704134828410.1089/1076327041348284. PMID 15265313. 
  52. ^ Fraser JK, Wulur I, Alfonso Z, Hedrick MH (April 2006). "Fat tissue: an underappreciated source of stem cells for biotechnology". Trends Biotechnol. 24 (4): 150'4. doi:10.1016/j.tibtech.2006.01.010. PMID 16488036. http://linkinghub.elsevier.com/retrieve/pii/S0167-7799(06)00028-X. 
  53. ^ Nakagami H, Morishita R, Maeda K, Kikuchi Y, Ogihara T, Kaneda Y (April 2006). "Adipose tissue-derived stromal cells as a novel option for regenerative cell therapy" ([dead link]). J. Atheroscler. Thromb. 13 (2): 77'81. PMID 16733294. http://joi.jlc.jst.go.jp/JST.JSTAGE/jat/13.77?from=PubMed. 
  54. ^ http://www.thehorse.com/ViewArticle.aspx?ID=13171
  55. ^ Black LL, Gaynor J, Adams C, et al. (2008). "Effect of intraarticular injection of autologous adipose-derived mesenchymal stem and regenerative cells on clinical signs of chronic osteoarthritis of the elbow joint in dogs". Vet. Ther. 9 (3): 192'200. PMID 19003780. 
  56. ^ Nixon AJ, Dahlgren LA, Haupt JL, Yeager AE, Ward DL (July 2008). "Effect of adipose-derived nucleated cell fractions on tendon repair in horses with collagenase-induced tendinitis". Am. J. Vet. Res. 69 (7): 928'37. doi:10.2460/ajvr.69.7.928. PMID 18593247. http://avmajournals.avma.org/doi/abs/10.2460/ajvr.69.7.928?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%3dncbi.nlm.nih.gov. 
  57. ^ PMID 16467274
  58. ^ PMID 16467274
  59. ^ PMID 19444176
  60. ^ PMID 16467274
  61. ^ [3]Stem Cell Treatment Institute
  62. ^ "Stem cells tailored to patients". BBC News. May 20, 2005. http://news.bbc.co.uk/2/hi/health/4555023.stm. Retrieved May 24, 2010. 
  63. ^ Hwang's Stem Cell Claims Further Discredited, http://news.sciencemag.org/sciencenow/2005/12/29-01.html


Related Articles & Resources

Sources Subject Index - Experts, Sources, Spokespersons

Sources Select Resources Articles







This article is based on one or more articles in Wikipedia, with modifications and additional content by SOURCES editors. This article is covered by a Creative Commons Attribution-Sharealike 3.0 License (CC-BY-SA) and the GNU Free Documentation License (GFDL). The remainder of the content of this website, except where otherwise indicated, is copyright SOURCES and may not be reproduced without written permission. (For information use the Contact form.)

SOURCES.COM is an online portal and directory for journalists, news media, researchers and anyone seeking experts, spokespersons, and reliable information resources. Use SOURCES.COM to find experts, media contacts, news releases, background information, scientists, officials, speakers, newsmakers, spokespeople, talk show guests, story ideas, research studies, databases, universities, associations and NGOs, businesses, government spokespeople. Indexing and search applications by Ulli Diemer and Chris DeFreitas.

For information about being included in SOURCES as a expert or spokesperson see the FAQ . For partnerships, content and applications, and domain name opportunities contact us.


Sources home page