Glossary Index | womenshealth.gov
By Dr. Matthew Watson
Find your glossary term by first letter:
a form of complementary and alternative medicine that involves inserting thin needles thorugh the skin at specific points on the body to control pain and other symptoms.
a form of complementary and alternative medicine that involves inserting thin needles thorugh the skin at specific points on the body to control pain and other symptoms.
written instructions letting others know the type of care you want if you are seriously ill or dying. These include a living will and health care power of attorney.
written instructions letting others know the type of care you want if you are seriously ill or dying. These include a living will and health care power of attorney.
disorders that involve an immune response in the body. Allergies are reactions to allergens such as plant pollen, other grasses and weeds, certain foods, rubber latex, insect bites, or certain drugs.
tiny glands in the breast that produce milk.
a brain disease that cripples the brain's nerve cells over time and destroys memory and learning. It usually starts in late middle age or old age and gets worse over time. Symptoms include loss of memory, confusion, problems in thinking, and changes in language, behavior, and personality.
clear, slightly yellowish liquid that surrounds the unborn baby (fetus) during pregnancy. It is contained in the amniotic sac.
when the amount of red blood cells or hemoglobin (the substance in the blood that carries oxygen to organs) becomes reduced, causing fatigue that can be severe.
Read the rest here:
Glossary Index | womenshealth.gov
Emerging interactions between skin stem cells and their …
By Dr. Matthew Watson
Hair follicle lineage and niche signals regulate hair follicle stem cells. (a) HFSCs can exist in two states. Quiescent bulge stem cells (Bu-SCs) are located in the outer layer of this niche and contribute to the generation of the outer root sheath. Primed stem cells reside in the hair germ, sandwiched between the bulge and a specialized dermal cluster known as the dermal papilla. They are responsible for generating the transit amplifying cell (TAC) matrix, which then gives rise to the hair shaft and its inner root sheath (IRS) channel. Although matrix and IRS are destroyed during catagen, many of the outer root sheath (ORS) cells are spared and generate a new bulge right next to the original one at the end of catagen. The upper ORS contributes to the outer layer of the new bulge, and the middle ORS contributes to the hair germ. Some of the lower ORS cells become the differentiated inner keratin 6+ (K6+) bulge cells, which provide inhibitory signals to Bu-SCs, raising their activation threshold for the next hair cycle. (b) During telogen, K6+ bulge cells produce BMP6 and FGF-18, dermal fibroblasts (DFs) produce BMP4 and subcutaneous adipocytes express BMP2. Together, these factors maintain Bu-SCs and hair germ in quiescence. At the transition to anagen, BMP2 and BMP4 are downregulated, whereas the expression of activation factors including noggin (NOG), FGF-7, FGF-10 and TGF-2 from dermal papillae and PDGF- from adipocyte precursor cells (APCs) is elevated. This, in turn, stimulates hair germ proliferation, and a new hair cycle is launched. Bu-SCs maintain their quiescent state until TAC matrix is generated and starts producing SHH.
Go here to read the rest:
Emerging interactions between skin stem cells and their ...
JCI – Bone marrow mesenchymal stem cells and TGF- …
By Dr. Matthew Watson
High levels of active TGF- in the bone marrow and abnormalities in bone remodeling are associated with multiple skeletal disorders. Genetic mutations in the TGF- signaling pathway cause premature activation of matrix latent TGF- and may manifest with various skeletal defects. There are additional diseases that result in high levels of active TGF-, which may contribute to the pathology. Here, we discuss how abnormal TGF- signaling results in uncoupled bone remodeling, mainly by loss of site-directed recruitment of MSCs that causes aberrant bone formation. Direct or indirect inhibition of TGF- signaling may provide potential therapeutic options for these disorders.
Genetic disorders. The critical role of TGF-1 in the reversal phase of bone remodeling is demonstrated by the range of skeletal disorders resulting from mutations in genes involved in TGF-1 signaling. Camurati-Engelmann disease (CED), characterized by a fusiform thickening of the diaphysis of the long bones and skull, is caused by mutations in TGFB1 that result in premature activation of TGF-1 (7174). Approximately 11 different TGFB1 mutations have been identified from families affected by CED (75, 76). All of the mutations are located in the region encoding LAP, either destabilizing LAP disulfide bridging or affecting secretion of the protein, both of which increase TGF-1 signaling, as confirmed by in vitro cell cultures and mouse models. Bone histology sections from patients with CED show decreased trabecular connectivity despite normal bone histomorphometric parameters with respect to osteoblast and osteoclast numbers (76, 77), suggestive of uncoupled bone remodeling. In vitro, the ratio of active to total TGF-1 in conditioned medium from cells expressing the CED mutant TGF-1 is significantly higher and enhances MSC migration (18). Targeted recruitment of MSCs to the bone-remodeling site is likely disrupted, secondary to loss of a TGF- gradient.
Elevations in TGF- signaling have also been observed in many genetic connective tissue disorders with craniofacial, skeletal, skin, and cardiovascular manifestations, including Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), and Shprintzen-Goldberg syndrome (SGS). MFS is caused by mutations in fibrillin and often results in aortic dilation, myopia, bone overgrowth, and joint laxity. Fibrillin is deposited in the ECM and normally binds TGF-, rendering it inactive. In MFS, the decreased level of fibrillin enhances TGF- activity (78). LDS is caused by inactivating mutations in genes encoding TRI and TRII (79). Physical manifestations include arterial aneurysms, hypertelorism, bifid uvula/cleft palate, and bone overgrowth resulting in arachnodactyly, joint laxity, and scoliosis. Pathologic analyses of affected tissue suggest chronically elevated TGF- signaling, despite the inactivating mutation (79). The mechanism of enhanced TGF- signaling remains under investigation. SGS is caused by mutations in the v-ski avian sarcoma viral oncogene homolog (SKI; refs. 80, 81) and causes physical features similar to those of MFS plus craniosynostosis. SKI negatively regulates SMAD-dependent TGF- signaling by impeding SMAD2 and SMAD3 activation, preventing nuclear translocation of the SMAD4 complex, and inhibiting TGF- target gene output by competing with p300/CBP for SMAD binding and recruiting transcriptional repressor proteins, such as mSin3A and HDACs (8284).
The neurocutaneous syndrome neurofibromatosis type 1 (NF1) has been noted to have skeletal features similar to those of CED, MFS, and LDS, including kyphoscoliosis, osteoporosis, and tibial pseudoarthrosis. Hyperactive TGF-1 signaling has been implicated as the primary factor underlying the pathophysiology of the osseous defects in Nf1fl/Col2.3Cre mice, a model of NF1 that closely recapitulates the skeletal abnormalities found in human disease (85). The exact mechanisms mediating mutant neurofibrominassociated enhancement of TGF- production and signaling remain unknown.
Osteoarthritis. While genetic disorders are rare, they have provided critical insight into the pathophysiology of more common disorders. Uncoupled bone remodeling accompanies the onset of osteoarthritis. TGF-1 is activated in subchondral bone in response to altered mechanical loading in an anterior cruciate ligament transection (ACLT) mouse model of osteoarthritis (86). High levels of active TGF-1 induced formation of nestin+ MSC clusters via activation of ALK5-SMAD2/3. MSCs underwent osteoblast differentiation in these clusters, leading to formation of marrow osteoid islets. Transgenic expression of active TGF-1 in osteoblastic cells alone was sufficient to induce osteoarthritis, whereas direct inhibition of TGF- activity in subchondral bone attenuated the degeneration of articular cartilage. Knockout of Tgfbr2 in nestin+ MSCs reduced osteoarthritis development after ACLT compared with wild-type mice, which confirmed that MSCs are the target cell population of TGF- signaling. High levels of active TGF-1 in subchondral bone likely disrupt the TGF- gradient and interfere with targeted migration of MSCs. Furthermore, mutations of ECM proteins that bind to latent TGF-s, such as small leucine-rich proteoglycans (87) and fibrillin (88), or mutations in genes involved in activation of TGF-, such as in CED (76) and LDS (89), are associated with high osteoarthritis incidence. Osteoblast differentiation of MSCs in aberrant locations appears histologically as subchondral bone osteoid islets and alters the thickness of the subchondral plate and calcified cartilage zone, changes known to be associated with osteoarthritis (90, 91). A computer-simulated model found that a minor increase in the size of the subchondral bone (1%2%) causes significant changes in the mechanical load properties on articular cartilage, which likely leads to degeneration (86). Importantly, inhibition of the TGF- signaling pathway delayed the development of osteoarthritis in both mouse and rat models (86).
MSCs in bone loss. Aging leads to deterioration of tissue and organ function. Skeletal aging is especially dramatic: bone loss in both women and men begins as early as the third decade, immediately after peak bone mass. Aging bone loss occurs when bone formation does not adequately compensate for osteoclast bone resorption during remodeling. Age-associated osteoporosis was previously believed to be due to a decline in survival and function of osteoblasts and osteoprogenitors; however, recent work by Park and colleagues found that mature osteoblasts and osteoprogenitors are actually nonreplicative cells and require constant replenishment from bone marrow MSCs (92). When MSCs fail to migrate to bone-resorptive sites or are unable to commit and differentiate into osteoblasts, new bone formation is impaired. Therefore, insufficient recruitment of MSCs, or their differentiation to osteoblasts, at the bone remodeling surface may contribute to the decline in bone formation in the elderly.
There are multiple hypotheses regarding the decreased osteogenic potential of MSCs during aging. For example, during aging, the bone marrow environment has an increased concentration of ROS and lipid oxidation that may decrease osteoblast differentiation, yet increase osteoclast activity (93, 94). MSCs also undergo senescence, which decreases proliferative capacity and contributes to decreased bone formation (95, 96). Cellular senescence involves the secretion of a plethora of factors, including TGF-, which induces expression of cyclin-dependent kinase inhibitors 2A and 2B (p16INK4A and p15INK4B, respectively; refs. 97).
Microgravity experienced by astronauts during spaceflight causes severe physiological alterations in the human body, including a 1%2% loss of bone mass every month during spaceflight (98). Several studies have shown decreases in osteoblastic markers of bone formation and increases in bone resorption (99101). The underlying molecular mechanisms responsible for the apparent concurrent decrease in bone formation and increase in bone resorption remain under investigation. Work by the McDonald group suggests that bone remodeling may become uncoupled under zero-gravity conditions secondary to decreased RhoA activity and resultant changes in actin stress fiber formation (102). In modeled microgravity, cultured human MSCs exhibit disruption of F-actin stress fibers within three hours of initiation of microgravity; the fibers are completely absent after seven days. RhoA activity is significantly reduced, and introduction of an adenoviral construct expressing constitutively active RhoA can reverse the elimination of stress fibers, significantly increasing markers of osteoblast differentiation (102). Under zero-gravity conditions, RhoA is unable to bind to its receptor, and a sufficient number of MSCs may not be able to migrate correctly to the bone-resorptive site for osteoblast differentiation, ultimately leading to bone loss with every cycle of remodeling.
Bone metastases are a frequent complication of cancer and often have both osteolytic and osteoblastic features, indicative of dysregulated bone remodeling. The importance of the bone marrow microenvironment contributing to the spread of cancer was first described in 1889 (103), postulating that tumor cells can grow only if they are in a conducive environment. Activation of matrix TGF- during bone remodeling plays a central role in the initiation of bone metastases and tumor expansion by regulating osteolytic and prometastatic factors (reviewed in refs. 104110). For example, TGF- can induce osteoclastic bone destruction by upregulating tumor cell expression of PTHrP and IL-11. Additionally, upregulation of CXCR4 by TGF- may home cancer cells to bones.
Originally posted here:
JCI - Bone marrow mesenchymal stem cells and TGF- ...
Peripheral-blood stem cells versus bone marrow from …
By Dr. Matthew Watson
See comment in PubMed Commons below N Engl J Med. 2012 Oct 18;367(16):1487-96. doi: 10.1056/NEJMoa1203517. Anasetti C, Logan BR, Lee SJ, Waller EK, Weisdorf DJ, Wingard JR, Cutler CS, Westervelt P, Woolfrey A, Couban S, Ehninger G, Johnston L, Maziarz RT, Pulsipher MA, Porter DL, Mineishi S, McCarty JM, Khan SP, Anderlini P, Bensinger WI, Leitman SF, Rowley SD, Bredeson C, Carter SL, Horowitz MM, Confer DL; Blood and Marrow Transplant Clinical Trials Network. Collaborators (182)
Horowitz MM, Carter SL, Confer DL, DiFronzo N, Wagner E, Merritt W, Wu R, Anasetti C, Logan BR, Lee SJ, Waller EK, Weisdorf DJ, Wingard JR, Couban S, Anderlini P, Bensinger WI, Leitman SF, Rowley SD, Carter SL, Karanes C, Horowitz MM, Confer DL, Allen C, Colby C, Gurgol C, Knust K, Foley A, King R, Mitchell P, Couban S, Pulsipher MA, Ehninger G, Johnston L, Khan SP, Maziarz RT, McCarty JM, Mineishi S, Porter DL, Bredeson C, Anasetti C, Lee S, Waller EK, Wingard JR, Cutler CS, Westervelt P, Woolfrey A, Logan BR, Carter SL, Lee SJ, Waller EK, Anasetti C, Logan BR, Lee SJ, Stadtmauer E, Wingard J, Vose J, Lazarus H, Cowan M, Wingard J, Westervelt P, Litzow M, Wu R, Geller N, Carter S, Confer D, Horowitz M, Poland N, Krance R, Carrum G, Agura E, Nademanee A, Sahdev I, Cutler C, Horwitz ME, Kurtzberg J, Waller EK, Woolfrey A, Rowley S, Brochstein J, Leber B, Wasi P, Roy J, Jansen J, Stiff PJ, Khan S, Devine S, Maziarz R, Nemecek E, Huebsch L, Couban S, McCarthy P, Johnston L, Shaughnessy P, Savoie L, Ball E, Vaughan W, Cowan M, Horn B, Wingard J, Silverman M, Abhyankar S, McGuirk J, Yanovich S, Ferrara J, Weisdorf D, Faber E Jr, Selby G, Rooms LM, Porter D, Agha M, Anderlini P, Lipton J, Pulsipher MA, Pulsipher MA, Shepherd J, Toze C, Kassim A, Frangoul H, McCarty J, Hurd D, DiPersio J, Westervelt P, Shenoy S, Agura E, Culler E, Axelrod F, Chambers L, Senaldi E, Nguyen KA, Engelman E, Hartzman R, Sutor L, Dickson L, Nademanee A, Khalife G, Lenes BA, Eames G, Sibley D, Gale P, Antin J, Ehninger G, Newberg NR, Gammon R, Montgomery M, Mair B, Rossmann S, Wada R, Waxman D, Ranlett R, Silverman M, Herzig G, Fried M, Atkinson E, Weitekamp L, Bigelow C, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Miller JP, Price T, Young C, Hilbert R, Oh D, Cable C, Smith JW, Kalmin ND, Schultheiss K, Beck T, Lankiewicz MW, Sharp D.
Randomized trials have shown that the transplantation of filgrastim-mobilized peripheral-blood stem cells from HLA-identical siblings accelerates engraftment but increases the risks of acute and chronic graft-versus-host disease (GVHD), as compared with the transplantation of bone marrow. Some studies have also shown that peripheral-blood stem cells are associated with a decreased rate of relapse and improved survival among recipients with high-risk leukemia.
We conducted a phase 3, multicenter, randomized trial of transplantation of peripheral-blood stem cells versus bone marrow from unrelated donors to compare 2-year survival probabilities with the use of an intention-to-treat analysis. Between March 2004 and September 2009, we enrolled 551 patients at 48 centers. Patients were randomly assigned in a 1:1 ratio to peripheral-blood stem-cell or bone marrow transplantation, stratified according to transplantation center and disease risk. The median follow-up of surviving patients was 36 months (interquartile range, 30 to 37).
The overall survival rate at 2 years in the peripheral-blood group was 51% (95% confidence interval [CI], 45 to 57), as compared with 46% (95% CI, 40 to 52) in the bone marrow group (P=0.29), with an absolute difference of 5 percentage points (95% CI, -3 to 14). The overall incidence of graft failure in the peripheral-blood group was 3% (95% CI, 1 to 5), versus 9% (95% CI, 6 to 13) in the bone marrow group (P=0.002). The incidence of chronic GVHD at 2 years in the peripheral-blood group was 53% (95% CI, 45 to 61), as compared with 41% (95% CI, 34 to 48) in the bone marrow group (P=0.01). There were no significant between-group differences in the incidence of acute GVHD or relapse.
We did not detect significant survival differences between peripheral-blood stem-cell and bone marrow transplantation from unrelated donors. Exploratory analyses of secondary end points indicated that peripheral-blood stem cells may reduce the risk of graft failure, whereas bone marrow may reduce the risk of chronic GVHD. (Funded by the National Heart, Lung, and Blood Institute-National Cancer Institute and others; ClinicalTrials.gov number, NCT00075816.).
Survival after Randomization in the Intention-to-Treat Analysis
The P value is from a stratified binomial comparison at the 2-year point. The P value from a stratified log-rank test was also not significant. A total of 75 patients in each group were still alive at 36 months.
N Engl J Med. 2012 October 18;367(16):10.1056/NEJMoa1203517.
Outcomes after Transplantation, According to Study Group
Continued here:
Peripheral-blood stem cells versus bone marrow from ...
Cancer – Wikipedia, the free encyclopedia
By Dr. Matthew Watson
Cancer i, also known as a malignant tumor or malignant neoplasm, is a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body.[1][2] Not all tumors are cancerous; benign tumors do not spread to other parts of the body.[2] Possible signs and symptoms include: a new lump, abnormal bleeding, a prolonged cough, unexplained weight loss, and a change in bowel movements among others.[3] While these symptoms may indicate cancer, they may also occur due to other issues.[3] There are over 100 different known cancers that affect humans.[2]
Tobacco use is the cause of about 22% of cancer deaths.[1] Another 10% is due to obesity, a poor diet, lack of physical activity, and consumption of ethanol (alcohol).[1] Other factors include certain infections, exposure to ionizing radiation, and environmental pollutants.[4] In the developing world nearly 20% of cancers are due to infections such as hepatitis B, hepatitis C, and human papillomavirus.[1] These factors act, at least partly, by changing the genes of a cell.[5] Typically many such genetic changes are required before cancer develops.[5] Approximately 510% of cancers are due to genetic defects inherited from a person's parents.[6] Cancer can be detected by certain signs and symptoms or screening tests.[1] It is then typically further investigated by medical imaging and confirmed by biopsy.[7]
Many cancers can be prevented by not smoking, maintaining a healthy weight, not drinking too much alcohol, eating plenty of vegetables, fruits and whole grains, being vaccinated against certain infectious diseases, not eating too much red meat, and avoiding too much exposure to sunlight.[8][9] Early detection through screening is useful for cervical and colorectal cancer.[10] The benefits of screening in breast cancer are controversial.[10][11] Cancer is often treated with some combination of radiation therapy, surgery, chemotherapy, and targeted therapy.[1][12] Pain and symptom management are an important part of care. Palliative care is particularly important in those with advanced disease.[1] The chance of survival depends on the type of cancer and extent of disease at the start of treatment.[5] In children under 15 at diagnosis the five year survival rate in the developed world is on average 80%.[13] For cancer in the United States the average five year survival rate is 66%.[14]
In 2012 about 14.1 million new cases of cancer occurred globally (not including skin cancer other than melanoma).[5] It caused about 8.2 million deaths or 14.6% of all human deaths.[5][15] The most common types of cancer in males are lung cancer, prostate cancer, colorectal cancer, and stomach cancer, and in females, the most common types are breast cancer, colorectal cancer, lung cancer, and cervical cancer.[5] If skin cancer other than melanoma were included in total new cancers each year it would account for around 40% of cases.[16][17] In children, acute lymphoblastic leukaemia and brain tumors are most common except in Africa where non-Hodgkin lymphoma occurs more often.[13] In 2012, about 165,000 children under 15 years of age were diagnosed with cancer. The risk of cancer increases significantly with age and many cancers occur more commonly in developed countries.[5] Rates are increasing as more people live to an old age and as lifestyle changes occur in the developing world.[18] The financial costs of cancer have been estimated at $1.16 trillion US dollars per year as of 2010.[19]
Cancers are a large family of diseases that involve abnormal cell growth with the potential to invade or spread to other parts of the body.[1][2] They form a subset of neoplasms. A neoplasm or tumor is a group of cells that have undergone unregulated growth, and will often form a mass or lump, but may be distributed diffusely.[20][21]
Six characteristics of cancer have been proposed:
The progression from normal cells to cells that can form a discernible mass to outright cancer involves multiple steps known as malignant progression.[22][23]
When cancer begins, it invariably produces no symptoms. Signs and symptoms only appear as the mass continues to grow or ulcerates. The findings that result depend on the type and location of the cancer. Few symptoms are specific, with many of them also frequently occurring in individuals who have other conditions. Cancer is the new "great imitator". Thus, it is not uncommon for people diagnosed with cancer to have been treated for other diseases, which were assumed to be causing their symptoms.[24]
Local symptoms may occur due to the mass of the tumor or its ulceration. For example, mass effects from lung cancer can cause blockage of the bronchus resulting in cough or pneumonia; esophageal cancer can cause narrowing of the esophagus, making it difficult or painful to swallow; and colorectal cancer may lead to narrowing or blockages in the bowel, resulting in changes in bowel habits. Masses in breasts or testicles may be easily felt. Ulceration can cause bleeding that, if it occurs in the lung, will lead to coughing up blood, in the bowels to anemia or rectal bleeding, in the bladder to blood in the urine, and in the uterus to vaginal bleeding. Although localized pain may occur in advanced cancer, the initial swelling is usually painless. Some cancers can cause a buildup of fluid within the chest or abdomen.[24]
General symptoms occur due to distant effects of the cancer that are not related to direct or metastatic spread. These may include: unintentional weight loss, fever, being excessively tired, and changes to the skin.[25]Hodgkin disease, leukemias, and cancers of the liver or kidney can cause a persistent fever of unknown origin.[24]
Read the rest here:
Cancer - Wikipedia, the free encyclopedia
japanese | StemCell Therapy MD
By Dr. Matthew Watson
SAN DIEGO(BUSINESS WIRE)Cytori Therapeutics, Inc. (NASDAQ: CYTX) today confirmed that two Japanese regenerative medicine laws, which went into effect on November 25, 2014, remove regulatory uncertainties and provide a clear path for the Company to commercialize and market Cytori Cell Therapy and its Celution System under the Companys existing and planned regulatory approvals.
Japans new regenerative medicine laws substantially clarify regulatory ambiguities of pre-existing guidelines and this news represents a significant event for Cytori, said Dr. Marc Hedrick, President & CEO of Cytori. We have a decade of operating experience in Japan and Cytori is nicely positioned to see an impact both on existing commercial efforts and on our longer-term efforts to obtain therapeutic claims and reimbursement for our products.
Under the two new laws, Cytori believes its Celution System and autologous adipose-derived regenerative cells (ADRCs) can be provided by physicians under current Class I device regulations and used under the lowest risk category (Tier 3) for many procedures with only the approval by accredited regenerative medicine committees and local agencies of the Ministry of Health, Labour and Welfare (MHLW). This regulatory framework is expected to streamline the approval and regulatory process and increase clinical use of Cytori Cell Therapy and the Celution System over the former regulations.
Before these new laws were enacted, the regulatory pathway for clinical use of regenerative cell therapy was one-size-fits-all, irrespective of the risk posed by certain cell types and approaches, said Dr. Hedrick. Now, Cytoris point-of-care Celution System can be transparently integrated into clinical use by providers under our Class I device status and the streamlined approval process granted to cell therapies that pose the lowest risk. Our technology is unique in that respect.
Cytoris Celution System Is in Lowest of Three Risk Categories
The Act on the Safety of Regenerative Medicines and an amendment of the 2013 Pharmaceutical Affairs Act (the PMD Act), collectively termed the Regenerative Medicine Laws, replace the Human Stem Cell Guidelines. Under the new laws, the cell types used in cell therapy and regenerative medicine are classified based on risk. Cell therapies using cells derived from embryonic, induced pluripotent, cultured, genetically altered, animal and allogeneic cells are considered higher risk (Tiers 1 and 2) and will undergo an approval pathway with greater and more stringent oversight due to the presumed higher risk to patients. Cytoris Celution System, which uses the patients own cells at the point-of-care, will be considered in the lowest risk category (Tier 3) for most cases, and will be considered in Tier 2 if used as a non-homologous therapy.
Streamlined Regulatory Approval for Certain Medical Devices
In the near future, Cytori intends to pursue disease-specific or therapeutic claims and reimbursement for Cytoris Celution System and the Company would, at that point, sponsor a clinical trial to obtain Class III device-based approval and reimbursement. The new laws include changes to streamline regulation of Class II and some Class III devices, which will now require the approval of certification bodies rather than the PMDA, similar to the European notified body model. To date, certification bodies have only been used for some Class II devices.
Conditional Regulatory Approval and Reimbursement Potential
As a supplementary benefit to Cytori, the Company may also choose to take advantage of the new conditional approval opportunities granted under the new laws. Once clinical safety and an indication of efficacy are shown, sponsors may apply for their cell product to receive conditional approval for up to seven years and may be eligible for reimbursement under Japans national insurance coverage. Under the conditional approval, the sponsor can then generate post-marketing data to demonstrate further efficacy and cost effectiveness.
More here:
japanese | StemCell Therapy MD
Stem Cells Advanced Skin Care | Introstem
By Dr. Matthew Watson
Human beings entirely regenerate their skin every 7 days. Cuts and wounds heal themselves and disappear from sight within a couple of weeks. Every cell within the skeleton is replaced within 7 years. This all goes to show how dynamic our cells really are. A number of medical experts have mentioned that the future of medicine lies in understanding how the body creates a single cell and the various mechanisms that are involved in renewing the cell throughout life. It is believed that once this goal is achieved, serious diseases such as Alzheimers, cancer, spinal cord injuries and diabetes can also be treated.
Medical science may have a long way to go when it comes to understanding stem cells, but the world of skin care has managed to achieve significant breakthroughs. Studies have shown the numerous benefits of adding stem cell technology into skin care products, and this has made stem cells one of the latest buzzwords in skin care. Stem cells have theamazing ability of being able to develop into different types of cells. When these cells divide, they can remain as the original stem cell or transform into another cell type, such as a skin cell.
Thus, stem cells are different from other types of cells for two simple reasons they can renew themselves and can also mimic other cells to serve specific functions. Their regenerative properties make them extremely crucial for skin care, as they offer a new way to look at anti-aging and treating things like lines, wrinkles and other aging signs.
One of the most interesting studies on the use of stem cells in skin care was conducted by Dr. Gregory Bays Brown, a former plastic surgeon. During the course of Dr. Browns research, it became evidently clear that a substance known as Epidermal Growth Factor was released whenever the body suffered from wounds or injury in order to accelerate the healing process. It has been believed that these same molecules can be used to regenerate aging skin by making stem cells mimic these factors.
Studies have also shown that stem-cell production decreases due to things like pollution and the damage caused by UV rays. In the year 2008, LVMH Laboratories identified certain key ingredients which had the ability to protect the stem cells from external factors. According to experts, the power of protecting stem cells was extremely vital for maintaining the youthful appearance of the skin and boosting epidermal regeneration.
Another exciting study surrounding the anti-aging effects of stem cells derived from apples was conducted by researchers working for Mibelle Biochemistry. They first obtained human stem cells to show that a minute concentration of 0.1% of these cells could stimulate the proliferation of stem cells within the body by as much as 80%! The researchers then conducted a second experiment where they irradiated the umbilical cord stem cells with UV light. About half of the stem cells that were cultured using growth mediums ended up dying, but the stem cells that were cultured using apple extracts showed a very small decrease. This samestudy also includedan experiment to observe the anti-wrinkle effects of stem cell potions created using apple extracts. This potion was applied on the crows feet area of 20 people, 2 times each day. After just two weeks, the wrinkle depth reduced by 8%. This decrease increased to 15% within 4 weeks, thereby causing a reduction in the overall signs of aging.
Better yet, stem cells havent just been influencing the world of skin care. The NeuralStem trial has already demonstrated that human embryonic stem cells can be transplanted into the spinal cord to help people suffering from ALS. Research on the same technique is underway to determine whether the treatment can slow thedecline of health or improve functioning in the body.
Although stem cell studies have a long way to go before their exact benefits are known, researchers believe that topical applications may stimulate the growth of new stem cells, thereby keeping the skin young and healthy.
Stem cell technology could lead to ailing heart mending …
By Dr. Matthew Watson
Tsai et al./Stem Cell Reports 2015
Weill Cornell investigators have discovered how to generate large numbers of rare cells in the network that pushes the heart's chambers to consistently contract. In this image, investigators stained these cells, generated from embryonic stem cells, to reveal cell-specific genes (green and red, indicated by arrows). The blue represents stained cell nuclei.
For the first time, scientists can efficiently generate large numbers of rare cells in the network that pushes the heart's chambers to consistently contract. The technique, published May 28 in Stem Cell Reports, could be a first step toward using a person's own cells to repair an irregular heartbeat known as cardiac arrhythmia.
This study, while done using mouse cells, will now allow us to develop human heart cells and test their function in repairing damaged hearts, said the study's senior author, Dr. Todd Evans, vice chair for research and the Peter I. Pressman Professor in the Department of Surgery at Weill Cornell Medical College.
The human heart beats billions of times during a lifetime, so it's not surprising that development of irregular heartbeats can lead to a variety of cardiac diseases, Evans says. But treatments for these disorders are costly, and often ineffective.
The government pays more than $3 billion each year for cardiac arrhythmia-related diseases. Despite this enormous expense, the treatments we have available are inadequate, Evans said. For example, artificial pacemakers are often used, but these can fail, and are particularly challenging therapies for children.
One solution is to coax a patient's own cells to generate the specific kinds of cells in the cardiac conduction system (CCS) that maintain a regular heartbeat.
We can imagine someday using these cells, for example, to create patches that can replace defective conduction fibers. Of course this is still a long way off, as we would need to study how to coax them into integrating properly with the rest of the CCS, Evans said. But previously, we did not even have the capacity to generate the cells, and now we can do so in a manner that is scalable, so that such preclinical research is now feasible.
Evans worked with Dr. Shuibing Chen, an expert in stem cell and chemical biology, and Dr. Su-Yi Tsai, a postdoctoral fellow and the study's lead investigator. Other key contributors were from the laboratory of Dr. Glenn Fishman, who specializes in cardiac physiology at New York University.
Their first goal was to increase the efficiency of coaxing mouse embryonic stem cells to become CCS cells. They created mouse stem cells that can express a CCS marker gene that can be quantified. This allows them to measure how many embryonic cells morph into CCS cells.
Read the rest here:
Stem cell technology could lead to ailing heart mending ...
Side Effects of Stem Cell/Bone Marrow Transplantation …
By Dr. Matthew Watson
Key Messages:
Although a stem cell transplant (sometimes called a bone marrow transplant) is an effective treatment for several types of cancer, it can cause a number of different side effects. The type and intensity of these side effects vary from person to person and depend on the kind of transplant performed, the person's overall health, and other factors. Your health care team will work with you to prevent side effects or manage any that occur. This is called palliative or supportive care and is an important part of your overall treatment plan. Be sure to talk with your health care team about any side effects you experience, including new symptoms or a change in symptoms.
The two most serious side effects of stem cell transplantation are infection and graft-versus-host disease.
Infection
The chemotherapy and/or radiation therapy given before a stem cell transplant weakens a persons immune system, lowering the bodys defenses against bacteria, viruses, and fungi. That means stem cell recipients are especially vulnerable to infection during this early period of treatment.
Although most people think the greatest risk of infection is from visitors or food, most infections that occur during the first few weeks after a transplant are caused by organisms that are already in the patient's lungs, sinuses, skin, and intestines. Fortunately, most of these infections are relatively easy to treat with antibiotics.
The reduced immunity of the early transplant period lasts about two weeks, after which the immune system is back to near full strength and can keep most common germs at bay without the help of medications. This is true for both autologous (AUTO) transplant recipients (who receive their own stem cells) and allogeneic (ALLO) transplant recipients (who receive stem cells from another person).
However, a risk of serious infection remains for ALLO transplant recipients because they are given anti-rejection drugs. These drugs suppress the immune system to prevent the body from rejecting the donors stem cells. However, this low immunity also leaves the body more at risk for infection. This risk increases when more anti-rejection drugs are needed. Your treatment team will work with you to prevent and manage infections.
Graft-versus-host disease
People who have an ALLO transplant are also at risk of developing a post-transplant illness called graft-versus-host disease (GVHD). It occurs when the transplanted stem cells recognize the patients body as foreign and attack it, causing inflammation. GVHD ranges from mild to life-threatening. AUTO transplant recipients do not face this risk because the transplanted stem cells come from their own bodies.
Read more:
Side Effects of Stem Cell/Bone Marrow Transplantation ...
Stem cell – ADULT STEM CELL THERAPY IS AVAILABLE NOW!
By Dr. Matthew Watson
Australia - New Zealand - Asia & Pacific Rim - China - Italy
The Foundation is a privately funded philanthropic (non profit) organization advising un-well people about how to gain access to Adult Stem Cell Therapy (ASCT). The Foundation is also promoting a plan to its members on how to prevent or limit the progression of degenerative diseases and other conditions. Degenerative disease is an escalating world problem that, if not controlled, could bankrupt our health systems.
A major objective of the Foundation is to highlight that people suffering from degenerative conditions now have the option of considering Adult Stem Cell Therapy. This therapy may improve quality of life for sufferers of Arthritis, MS, Parkinsons, Diabetes, Stroke, Alzheimers, Spinal Cord injuries, Cancer or Chronic Pain to name a few. A stem cell transplant, instead of a joint replacement, is fast becoming the preferred first option for orthopedic surgeons.
The Foundation intends to educate parents/carers of children suffering from a debilitating or degenerative condition like Cerebral Palsy, Muscular Dystrophy, Autism, Spinal injuries, Cystic fibrosis, ADHD etc. Stem cell treatments have progressed in leaps and bounds for these conditions. There are now state of the art clinics that specialize in treating the afore-mentioned conditions. Children can usually benefit substantially from an early intervention by stem cell therapies and other protocols because they are still growing. As an example: spending time in a mild hyperbaric chamber (HBO) can also be beneficial. Just fill out the Application Form for an experimental transplant and we will be only too happy to advise.
The ASCF has become a global Information Centre for stem cell therapy. The centre will only support clinics that have demonstrated they abide by the highest medical standards and have a proven track record of administering these types of therapies, in Australia and overseas. We can now advise locally which gives peace of mind to our members who are contemplating a procedure of this nature.
Creating awareness of the availability of stem cell therapy and that it has become viable for consideration.
To raise money from benefactors, including private and commercial sponsorships.
Visit link:
Stem cell - ADULT STEM CELL THERAPY IS AVAILABLE NOW!
Repairing Chronic Muscle Tears with Stem Cells – Video
By Dr. Matthew Watson
Repairing Chronic Muscle Tears with Stem Cells
Chronic muscle tears like hamstring pulls and shoulder rotator cuff muscles are tough to heal. Research suggests that injecting bone marrow stem cells into the area may solve that problem.
By: Chris Centeno
See the article here:
Repairing Chronic Muscle Tears with Stem Cells - Video
Stem Cells for Skin Tissue Engineering and Wound Healing
By Dr. Matthew Watson
1. Keratinocytes
There are two approaches to commit ES cells and adult stem cells (of non-epidermal origin) to the keratinocyte lineage in vitro. One approach would be to expose the cells to a cocktail of exogenous cytokines, growth factors, chemicals, and extracellular matrix (ECM) substrata over a prolonged duration of in vitro culture. Only a fraction of the stem cells would be expected to undergo commitment to the keratinocyte lineage, because many of these cytokines, growth factors, chemicals, and ECM substrata would exert non-specific pleitropic effects on stem cell differentiation into multiple lineages. At best, the cocktail combination of various cytokines, growth factors, chemicals, and ECM substrata can be optimized by trial and error, to maximize the proportion of stem cells committing to the keratinocyte lineage, while at the same time yielding a large number of other undesired lineages. Hence, extensive selection/purification and proliferation of the commited keratinocyte progenitors is likely to be required.
By using such an approach, Coraux et al.54 managed to achieve commitment and subsequent differentiation of murine ES cells into the keratinocyte lineage, in the presence of a cocktail combination of bone morphogenetic protein-4 (BMP-4), ascorbate, and ECM derived from human normal fibroblasts (HNFs) and murine NIH-3T3 fibroblasts. Nevertheless, it must be noted that the study of Coraux et al.54 also reported a high degree (approximately 80%) of non-specific differentiation into multiple uncharacterized lineages, and no attempt was made to purify differentiated keratinocytes or keratinocyte progenitors from the mixture of lineages derived from murine ES cells. Bagutti et al.61 reported that coculture with human dermal fibroblasts (HDFs) as well as HDF-conditioned media could induce beta integrin- deficient murine ES cells to commit and differentiate into the keratinocyte lineage. However, as with the study of Coraux et al.,54 the keratinocytes were interspersed with differentiated cells of other lineages. Recently, differentiation of human ES cells into the keratinocyte lineage was also reported by Green et al.62 However, this study was based on in vivo teratoma formation within a SCID mouse model, and to date, there are no parallel in vitro studies that have been reported.
With adult stem cells of non-epidermal origin, there are also few studies 63, 64 which have successfully achieved re-commitment and trans-differentiation to the keratinocyte lineage. Even so, these studies were based primarily on the transplantation of undifferentiated stem cells in vivo, with the observed trans-differentiation occurring sporadically and at extremely low frequencies. Moreover, the validity of the experimental data may be clouded by controversy over the artifact of stem cell fusion in vivo.65 To date, there are no parallel in vitro studies that have achieved recommitment and trans-differentiation of non-epidermal adult stem cells to the keratinocyte lineage. It can therefore be surmised that the use of exogenous cytokines, growth factors, chemicals, and ECM substrata to induce ES cell and nonepidermal adult stem cell commitment to the keratinocyte lineage is a relatively inefficient, time-consuming, and labor-intensive process that would require extensive selection and purification of the committed keratinocyte progenitors. Hence, it would be technically challenging to apply this to the clinical situation.
The other approach for inducing ES cell and non-epidermal adult stem cell commitment to the keratinocyte lineage is through genetic modulation. This may be achieved by transfecting stem cells with recombinant DNA constructs encoding for the expression of signaling proteins that promote commitment to the keratinocyte lineage. Of particular interest are the Lef-1/Tcf family of Wnt regulated transcription factors that act in concert with b-catenin,66, 67 c-myc which is a downstream target of the Wnt-signaling pathway,68, 69 and the transactivation domain containing isoform of transcription factor p63 (Tap63).70, 71 Interestingly, the transcription factor GATA-3, which is well known to be a key regulator of T-cell lineage determination, has also been shown to be essential for stem cell lineage determination in skin, where it is expressed at the onset of epidermal stratification and Inner Root Sheath (IRS) specification in follicles.72 Recombinant overexpression of p6373 and c-Myc74 has been reported to promote commitment and differentiation to the keratinocyte lineage.
The disadvantage of directing differentiation through genetic modulation is the potential risks associated with utilizing recombinant DNA technology in human clinical therapy. For example, the overexpression of any one particular protein within transfected stem cells would certainly have unpredictable physiological effects upon transplantation in vivo. This problem may be overcome by placing the recombinant expression of the particular protein under the control of switchable promoters, several of which have been developed for expression in eukaryotic systems. Such switchable promoters could be responsive to exogenous chemicals,75 heat shock,76 or even light.77 Genetically modified stem cells may also run the risk of becoming malignant within the transplanted recipient. Moreover, there are overriding safety concerns with regard to the use of recombinant viral based vectors in the genetic manipulation of stem cells.78 It remains uncertain as to whether legislation would ultimately permit the use of genetically modified stem cells for human clinical therapy. At present, the potential detrimental effects of transplanting genetically modified stem cells in vivo are not well studied. More research needs to be carried out on animal models to address the safety aspects of such an approach.
More recently, there is emerging evidence that some transcription factors (which are commonly thought of as cytosolic proteins) have the ability to function as paracrine cell to cell signaling molecules.79 This is based on intercellular transfer of transcription factors through atypical secretion and internalization pathways.79 Hence, there is an exciting possibility that transcription factors implicated in commitment to the keratinocyte lineage may in the future be genetically engineered to incorporate domains that enable them to participate in novel paracrine signaling mechanisms. This in turn would have tremendous potential for inducing the commitment of ES cells and non-epidermal adult stem cells to the keratinocyte lineage.
Skin appendages, including hair follicles, sebaceous glands and sweat glands, are linked to the epidermis but project deep into the dermal layer. The skin epidermis and its appendages provide a protective barrier that is impermeable to harmful microbes and also prevents dehydration. To perform their functions while being confronted with the physicochemical traumas of the environment, these tissues undergo continual rejuvenation through homeostasis, and, in addition, they must be primed to undergo wound repair in response to injury. The skins elixir for maintaining tissue homeostasis, regenerating hair, and repairing the epidermis after injury is its stem cells.
The hair follicle is composed of an outer root sheath that is contiguous with the epidermis, an inner root sheath and the hair shaft. The matrix surrounding the dermal papilla, in the hair root, contains actively dividing, relatively undifferentiated cells and is therefore a pocket of MSCs that are essential for follicle formation. The lower segment of each hair follicle cycles through periods of active growth (anagen), destruction (catagen) and quiescence (telogen).80 A specialized region of the outer root sheath of the hair follicle, known as the bulge, is located below the sebaceous gland, which is also the attachment site of the arrector pili muscle, receiving inputs from sensory nerve endings and blood vessels. Furthermore, the hair follicle bulge is a reservoir of slow-cycling multipotent stem cells.81, 82 Subsets of these follicle-derived multipotent stem cells can be activated and migrate out of hair follicles to the site of a wound to repair the damaged epithelium; however, they contribute little to the intact epidermis. These hair follicle stem cells can also contribute to the growth of follicles themselves and the sebaceous gland. For example, in the absence of hair follicle stem cells, hair follicle and sebaceous gland morphogenesis is blocked, and epidermal wound repair is compromised.83 In addition to containing follicle epidermal stem cells, the bulge contains melanocyte stem cells.84 Recent studies show that nestin, a marker for neural progenitor cells, is selectively expressed in cells of the hair follicle bulge and that these stem cells can differentiate into neurons,85 glia, keratinocytes, smooth muscle cells, melanocytes and even blood vessels.86, 87 Examination of close developmental and anatomical parallels between epithelial tissue and dermal tissue in skin and hair follicles has revealed dermal tissue to have stem cells. Paus et al. indicated that hair follicle dermal sheath cells might represent a source of dermal stem cells that not only incorporate into the hair-supporting papilla, low down in the follicle, but also move up and out from the follicle dermal sheath into the dermis of adjoining skin.88 Hair follicle dermal sheath cells taken from the human scalp can form new dermal papilla, induce the formation of hair follicles, and produce hair shafts when transplanted onto skin.89 There is also a clear transition from dermal sheath to dermal papilla cells.90 When the follicle dermal cells are implanted into skin wounds, they can be incorporated into the new dermis in a manner similar to that of skin wound-healing fibroblasts.91 However, these cell populations still lack specific markers for purifying and distinguishing the stem cells from their progeny. Furthermore, of prime importance is improving our understanding of the relation between bulge cells and interfollicular epidermal stem cells and between bulge cells and other stem cells inhabiting the skin and the mechanisms of hair growth.
Recently, cell replacement therapy has offered a novel and powerful medical technology for skin repair and regeneration: a new population of stem cell, called a neural crest stem cell, from adult hair follicles, was discovered to have the ability to differentiate in vitro to keratinocytes, neurons, cartilage/bone cells, smooth muscle cells, melanocytes, glial cells, and adipocytes.9296 In mammalian skin, skin-derived neural progenitors were isolated and expanded from the dermis of rodent skin and adult human scalp and could differentiate into both neural and mesodermal progeny.97, 98 Skin-derived neural progenitor cells were isolated based on the sphere formation of floating cells after 37 days of culture in uncoated flasks with epidermal growth factor and fibroblast growth factor, and characterized by the production of nestin and fibronectin, markers of neural precursors. In addition, skin-derived neural progenitor cells were identified as neural crest derived by the use of Wnt1 promoter driving LacZ expression in the mouse. Some of the LacZ-positive cells were found in the skin of the face, as well as in the dermis and dermal papilla of murine whisker.99 These skin derived neural crest cells have already shown promising results in regenerative medicine such as the promotion of regenerative axonal growth after transplantation into injured adult mouse sciatic nerves 95 or spinal cord repair,100 resulting in the recovery of peripheral nerve function. This new study marks an important first step in the development of real stem-cell-based therapies and skin tissue regeneration.
More here:
Stem Cells for Skin Tissue Engineering and Wound Healing
What Happens During My Stem Cell Therapy Procedure? – Video
By Dr. Matthew Watson
What Happens During My Stem Cell Therapy Procedure?
Ever wonder what happens during your stem cell therapy procedure? This video describes the process step-by-step with Orlando Orthopaedic Center #39;s Dr. G. Grady McBride. For more visit http://www.
By: OrlandoOrtho
Link:
What Happens During My Stem Cell Therapy Procedure? - Video
Scientists develop cardiac cells using stem cells
By Dr. Matthew Watson
For millions of people around the world, who suffer from various diseases, research in stem cells offers a ray of hope. Scientists of the city-based Indian Institute of Science have used stem cells of a mouse to culture cardiac cells.
Explaining the research, Polani B. Seshagiri said their research over the past seven years has helped develop cardiac cells that function and beat in rhythms identical to the original cell.
Speaking on Stem Cell Awareness Day recently, Prof. Seshagiri said stem cells had several advantages and could cure human disorders and diseases, which could not be cured by conventional approaches. However, he warned that there was a need to be aware of the limitations of stem cells.
Sudarshan Ballal, Medical Director, Manipal Health Enterprise, said stem cells had enormous potential as they never die and could be converted into any cell. Stem cells can be converted into organs and maybe years later, organs can be cultivated in labs through stem cell, he said. Elaborating further, he said a stem cell could be compared to a bicycle, which could turn into car, motorbike and spaceship based on the environment and conditions.
Nazeer Ahmed, Deputy Drug Controller of Karnataka, said they were in the process of chalking out regulations for stem cells as there were currently no rules to regulate stem cell research and therapy.
Please Wait while comments are loading...
1. Comments will be moderated by The Hindu editorial team. 2. Comments that are abusive, personal, incendiary or irrelevant cannot be published. 3. Please write complete sentences. Do not type comments in all capital letters, or in all lower case letters, or using abbreviated text. (example: u cannot substitute for you, d is not 'the', n is not 'and'). 4. We may remove hyperlinks within comments. 5. Please use a genuine email ID and provide your name, to avoid rejection.
Read more:
Scientists develop cardiac cells using stem cells
MaryannAstle published Tried & Tested: Best beauty products new to the market
By Dr. Matthew Watson
Comments(0)
From cleansers and toners to salt scrubs and perfumes there's plenty of beauty treats that have just been released. Mary-ann Astle puts forward some of the best new releases on the beauty market....
NURISS Swiss Apple Stem Cell Rejuvenator Serum
Skincare and wellness brand Nuriss has a new star product in the making. The Swiss Apple Stem Cell Rejuvenator Serum (30ml, 120) uses the longevity found in stem cells of the rare species of Swiss apple (the Uttwiler Sptlauber) to repair and rejuvenate your skin. When applied to the skin it can help with wrinkle reduction and increase collagen production.
Without wanting to blind you with science the serum is created by cultivating the apple's stem cells which are rich in phytonutrients and proteins which are beneficial to human skin. You don't need to use a lot to see the benefits after cleansing and toning, smooth one or two drops over your face and neck. Use morning and night to get the best results.
Click here to go to Nuriss
LouLouBelle Skincare of London
LouLouBelle has a new range of skincare products that will not only pamper you but which also smell absolutely gorgeous.
With tantalising blends like Geranium and Tea Tree, Lavender and Cypress and Palmarosa and Patchouli, LouLouBelle London is a boutique aromatherapy brand that uses natural ingredients to help make your skin feel great and smell delightful. It's also reasonably priced with cleansers (200ml, 19.95), toners (150ml, 17.95) and moisturisers (50ml, 24.95).
Every product is formulated from its own unique recipe that is created by selecting essential oils, plant essences and floral waters to match the specific requirements of a given skin type. The result is a refreshing range of cleansers, toners and moisturisers that are available in a different blend for each of the three main categories of skin dry skin, combination skin and problem/oily skin.
More here:
MaryannAstle published Tried & Tested: Best beauty products new to the market
Human 'heart on a chip' to aid drug tests
By Dr. Matthew Watson
for the first time, scientists - including an Indian American bioengineer - have developed a network of pulsating cardiac muscle cells housed in an inch-long silicone device that effectively models human heart tissue.
This organ-on-a-chip represents a major step forward in the development of accurate, faster methods of testing for drug toxicity.
"Ultimately, these chips could replace the use of animals to screen drugs for safety and efficacy," said professor Kevin Healy of University of California, Berkeley, who led the team.
"This system is not a simple cell culture where tissue is being bathed in a static bath of liquid," said study lead author Anurag Mathur, a postdoctoral scholar in Healy's lab.
"We designed this system so that it is dynamic. It replicates how tissue in our bodies actually gets exposed to nutrients and drugs," Mathur explained.
The study authors noted a high failure rate associated with the use of nonhuman animal models to predict human reactions to new drugs.
Much of the failure is due to fundamental differences in biology between species, the researchers explained.
"Using a well-designed model of a human organ could significantly cut the cost and time of bringing a new drug to market," Healy added.
The heart cells were derived from human-induced pluripotent stem cells, the adult stem cells that can be coaxed to become many different types of tissue.
The researchers designed their cardiac microphysiological system, or heart-on-a-chip, so that its 3D structure would be comparable to the geometry and spacing of connective tissue fibre in a human heart.
Read the original here:
Human 'heart on a chip' to aid drug tests
Dr. Raj at Beverly Hills Orthopedic Institute Now Offering Stem Cell Therapy to Heal Chronic Tendonitis
By Dr. Matthew Watson
Beverly Hills, California (PRWEB) April 07, 2015
Dr. Raj, the top Orthopedic Surgeon in Beverly Hills and Los Angeles, is now offering stem cell therapy to heal chronic tendonitis. The treatment works exceptionally well for those suffering from tendonitis of the rotator cuff, achilles, elbow and knee. For more information and scheduling, call (310) 247-0466.
As a pioneer in regenerative medicine, Dr. Raj has been helping patients with degenerative arthritis achieve relief and avoid joint replacements for years with stem cell procedures. By adding the procedures for tendonitis, Dr. Raj is now helping patients avoid potentially risky surgeries and get back to being more active for soft tissue related pain.
"Surgery for tendonitis is often not 100% successful for patients, and the rehabilitation period may take six months," states Dr. Raj. "With the stem cell therapy, pain relief is quick and athletes get back to sports faster!"
Regenerative medicine for tennis elbow has been shown in research studies to be effective at relief and helping avoid surgery. A 2013 study out of South Florida showed that 28 out of 30 patients with chronic tennis elbow avoided surgery and got back to being very active.
For several years in a row, Dr. Raj has been named the top orthopedic doctor in Los Angeles and Beverly Hills. He is an ABC News Medical Correspondent as well as a WebMD Medical Expert.
Hundreds of patients have benefited from stem cell procedures with Dr. Raj at Beverly Hills Orthopedic Institute. They come from all over Southern California, along with throughout the country. Call (310) 247-0466 for scheduling stem cell therapy with an orthopedic surgeon Beverly Hills trusts and respects.
See original here:
Dr. Raj at Beverly Hills Orthopedic Institute Now Offering Stem Cell Therapy to Heal Chronic Tendonitis
Is a loophole in stem cell law helping new therapy to thrive, or allowing dubious science?
By Dr. Matthew Watson
Life-changing results: Sandra Sharman is a private stem cell patient. Photo: Meredith O'Shea
Last week, Suzie Palmer, 44, travelled from her home in NSW to the Gold Coast for her second round of stem cell treatments for multiple sclerosis. OnTuesday morning,the wheelchair-bound poet underwent liposuction.
By 2.30pm, stem cells had been partially separated from her abdominal fat, suspended in plasma, and injected intravenously. Her doctor, Soraya Felix, is a cosmetic surgeon and molecular biologist with a sideline in regenerative medicine.
Palmer, a relentlessly upbeat and positive person, says the treatments have helped her cope better with heat, improved her mobility and flexibility and otherwise made her "feel like a normal human being". She has, she says, managed a few steps with a walker, still a long way from "running about, which is my dream".
Poster girl: Suzie Palmer is undergoing stem cell therapy for MS. Photo: Edwina Pickles
The rapidly growing stem cell industry is aglow with similarly positive testimonials, notably on behalf of practitioners who offer little documented scientific evidence of their success.
Advertisement
Suzie Palmer is literally the poster girl for stem cell tourism within Australia. You can find her smiling sweetly, along with Dr Felix, on the Facebook page of a group called the Adult Stem Cell Foundation. She is one of an unknown number of unwell Australians pinning their hopes on an unregulated industry that is now under review by the Therapeutic Goods Administration.
The TGA public consultation, which closed earlier this month, was prompted by long-standing concerns raised by Stem Cells Australia that a loophole in the regulations has allowed dozens of doctors across Australia to provide experimental treatments without the ethics committee oversight that registered clinical trials are subject to. These treatments invariably cost $10,000 and up. The loophole is this: while the use of donor stem cells in therapies is tightly regulated, the use of a patient's own stem cells is not.
Professor Martin Pera is the program leader of Stem Cells Australia, which is administered by the University of Melbourne and includes scientists from Monash University, the Walter and Eliza Hall Institute for Medical Research, the Florey Institute and the CSIRO, among others. They are engaged in a seven-year Australian Research Council project to answer the big questions about stem cells and the potential for reliable therapies.
Read more from the original source:
Is a loophole in stem cell law helping new therapy to thrive, or allowing dubious science?
Induced Pluripotent Stem Cell (iPSC) Industry Complete Report 2015 – 2016
By Dr. Matthew Watson
DALLAS, April 2, 2015 /PRNewswire/ --
Lifescienceindustryresearch.com adds "Complete 2015-16 Induced Pluripotent Stem Cell (iPSC) Industry Report" in its store. Recent months have seen the first iPSC clinical trial in humans, creation of the world's largest iPSC Biobank, major funding awards, a historic challenge to the "Yamanaka Patent", a Supreme Court ruling affecting industry patent rights, the announcement of an iPSC cellular therapy clinic scheduled to open in 2019, and much more. Furthermore, iPSC patent dominance continues to cluster in specific geographic regions, while clinical trial and scientific publication trends give clear indicators of what may happen in the industry in 2015 and beyond.
Is it worth it to get informed about rapidly-evolving market conditions and identify key industry trends that will give an advantage over the competition?
BrowsetheReportComplete 2015-16 Induced Pluripotent Stem Cell (iPSC) Industry Reportathttp://www.lifescienceindustryresearch.com/complete-2013-14-induced-pl ....
Induced pluripotent stem cells represent a promising tool for use in the reversal and repair of many previously incurable diseases. The cell type represents one of the most promising advances discovered within the field of stem cell research during the past decade, making this a valuable industry report for both companies and investors to claim in order to optimally position themselves to sell iPSC products. To profit from this lucrative and rapidly expanding market, you need to understand your key strengths relative to the competition, intelligently position your products to fill gaps in the market place, and take advantage of crucial iPSC trends.
Report Applications
This global strategic report is produced for: Management of Stem Cell Product Companies, Management of Stem Cell Therapy Companies, Stem Cell Industry Investors
It is designed to increase your efficiency and effectiveness in:
Four Primary Areas of Commercialization
There are currently four major areas of commercialization for induced pluripotent stem cells, as described below:
Read the original post:
Induced Pluripotent Stem Cell (iPSC) Industry Complete Report 2015 - 2016
Blood ties: Ky. basketball fan gets Wisconsin assist
By Dr. Matthew Watson
Scott Logdon is a die-hard University of Kentucky basketball fan, but he can't deny he's got some Wisconsin blood in him -- literally.
When the father of four was being treated for high-risk leukemia at UK in 2013, 20-year-old University of Wisconsin student Chris Wirz anonymously donated bone marrow stem cells to him. The two men first spoke just after the Wildcats bested the Badgers during last year's NCAA Final Four, and basketball was a frequent topic of conversation as their friendship grew.
While each will be rooting for his own team during this Saturday's Final Four rematch, both say they have a soft spot for the other team.
"I've stayed true to UK," said Logdon, 44, of Salvisa, Ky. "But when I talked to Chris for the first time I told him, 'That's why I felt so bad when we beat you: I've got Badger blood in me!"'
Wirz, who lives three blocks from where the Badgers play, hopes Wisconsin wins this year, and has even predicted an upset in his basketball bracket. "Who doesn't want to root for the underdog?" he said.
But he plans to send a text of congratulations if Logdon's team wins -- since their connection is much deeper than basketball rivalry.
"We're literally working off the same immune system," said Wirz, now 22 and a University of Wisconsin senior. "This has been one of the most emotionally overwhelming experiences of my life, realizing how important he is to his family and his community and seeing the hole that would've been left by him."
A dire diagnosis
Logdon, chief deputy at Woodford County Detention Center in Versailles, Ky., and a youth minister in his church, recalled playing basketball with teenagers just a few nights before going to the doctor for what his wife, Angela, initially thought was strep.
But tests showed he had acute myeloid leukemia, a blood cancer estimated by the American Cancer Society to have stricken 18,860 Americans last year and killed about 10,460, mostly adults.
Excerpt from:
Blood ties: Ky. basketball fan gets Wisconsin assist