Eli and Edythe Broad Center for Regenerative Medicine and …

Welcome

Welcome to the Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, located on the University of Southern Californias Health Sciences Campus.

Our investigators are exploring the normal mechanisms that build, maintain and repair our body systems, to develop knowledge-based approaches for regenerative medicine. Scientists are researching kidney, liver, neural, blood, cardiovascular, skeletal and skin disease models.

The center serves as a hub for USC Stem Cell, which connects researchers in stem cell biology and regenerative medicine across USC.

Oct 9, 2015

As a winner of the NIH Directors New Innovator Award, USC Stem Cell principal investigator Min Yu will strive to develop individualized medicine targeting rare and deadly breast cancer stem cells. The five-year, $2.475 million award is part of the High-Risk, High-Reward Research program supported by the NIH Common Fund.

Sep 22, 2015

How do you turn stem cells into nephrons, the functional unit of the kidney? Albert D. Kim, PhD, a postdoctoral fellow in the laboratory of Andy McMahon, PhD, is exploring this question with support from a Hearst Fellowship, an award recognizing an exceptional junior postdoctoral fellow pursuing stem cell research at USC.

Sep 21, 2015

Once the stuff of science fiction, genetic engineering is now offered on a fee-for-service basis at USC. On September 19, USC Stem Cell faculty and staff welcomed their supporters, the Chang and Choi families, and nearly 100 of their friends to celebrate the grand opening of the Chang Stem Cell Engineering Facility, located on the second floor of the Eli and Edythe Broad Center (BCC) for Regenerative Medicine and Stem Cell Research at USC on the Health Sciences Campus. Established with a generous gift from the Chang family, the stem cell engineering facility will serve researchers at USC as well as at other institutions.

See the rest here:
Eli and Edythe Broad Center for Regenerative Medicine and ...

My new ‘neighbor’ in Sacramento: a fat stem cell clinic …

For years Ive been writing about stem cell clinics that sell non-FDA approved stem cell treatments to vulnerable patients right here in America.

These clinics have been sprouting up like mushrooms across the US and their numbers may be above200 today overall. As a result perhaps it was inevitable that one would arrive in a locale near me.

Tomorrow, July 11, reportedly the Irvine Stem Cell Treatment Center will open a Sacramento, CA branch.The doctor there will apparently be Thomas A. Gionis (picture from press release). This private, for-profit clinic has no affiliation with UC Davis School of Medicine in Sacramento where Im located.

The stem cell clinic Sacramento branch will sell transplants of fat stem cells in the form of something called stromal vascular fraction or SVF, which I believe is almost certainly a drug. To my knowledge this clinic and the large chain that it belongs to called Cell Surgical Network (CSN), do not have FDA approval to use SVF.

Both publicly and to me on this blog, CSN continues to arguethat it doesnt need FDA approval (here,hereandhere), but recent FDA draft guidances sure suggest otherwise in my view. Of course if the FDA never takes action on the use of SVF then how are we all supposed to interpret that? WithoutFDA action or finalized guidelines, is it formally possible that the FDA could back down on SVF?

This clinic will reportedly sell SVF to treat a dizzying array of conditions having nothing to do with fat:

Emphysema, COPD, Asthma, Heart Failure, Heart Attack, Parkinsons Disease, Stroke, Traumatic Brain Injury, Lou Gehrigs Disease, Multiple Sclerosis, Lupus, Rheumatoid Arthritis, Crohns Disease, Muscular Dystrophy, Inflammatory Myopathies, and Degenerative Orthopedic Joint Conditions (Knee, Shoulder, Hip, Spine).

To me as a scientist the use of SVF to treat all these very different conditions does not make good common sense.

It would also seem arguably to be quite likely be considered non-homologous use by the FDA, a standing that would also automatically make this a drug requiring FDA pre-approval. Non-homologous use means using a biological product of a certain kind that is not homologous (not the same or similar in origin) to the tissue being treated. For example, fat is not the same as the brain or other central nervous system tissue that is involved in several of the conditions on the clinic menu. Same goes for cardiac muscle, airways, etc.

The use of a non-FDA approved product in a largely non-homologous manner increases risks for patients. Note that these stem cell transplants are also very expensive with little evidence in the way of published data of benefit.

The CSN stem cell clinic in Sacramento will be located at the New Body MD Surgical Center, just about 10 minutes from my office. I plan on paying them a visit at some point. Lets see how that goes. Will they let me in?

. Bookmark the

.

Follow this link:
My new 'neighbor' in Sacramento: a fat stem cell clinic ...

Stem-cell therapy – Wikipedia, the free encyclopedia

This article is about the medical therapy. For the cell type, see Stem cell.

Stem-cell therapy is the use of stem cells to treat or prevent a disease or condition.

Bone marrow transplant is the most widely used stem-cell therapy, but some therapies derived from umbilical cord blood are also in use. Research is underway to develop various sources for stem cells, and to apply stem-cell treatments for neurodegenerative diseases and conditions, diabetes, heart disease, and other conditions.

With the ability of scientists to isolate and culture embryonic stem cells, and with scientists' growing ability to create stem cells using somatic cell nuclear transfer and techniques to create induced pluripotent stem cells, controversy has crept in, both related to abortion politics and to human cloning. Additionally, efforts to market treatments based on transplant of stored umbilical cord blood have been controversial.

For over 30 years, bone marrow has been used to treat cancer patients with conditions such as leukaemia and lymphoma; this is the only form of stem-cell therapy that is widely practiced.[1][2][3] During chemotherapy, most growing cells are killed by the cytotoxic agents. These agents, however, cannot discriminate between the leukaemia 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. The transplanted cells also generate an immune response that helps to kill off the cancer cells; this process can go too far, however, leading to graft vs host disease, the most serious side effect of this treatment.[4]

Another stem-cell therapy called Prochymal, was conditionally approved in Canada in 2012 for the management of acute graft-vs-host disease in children who are unresponsive to steroids.[5] It is an allogenic stem therapy based on mesenchymal stem cells (MSCs) derived from the bone marrow of adult donors. MSCs are purified from the marrow, cultured and packaged, with up to 10,000 doses derived from a single donor. The doses are stored frozen until needed.[6]

The FDA has approved five hematopoietic stem-cell products derived from umbilical cord blood, for the treatment of blood and immunological diseases.[7]

In 2014, the European Medicines Agency recommended approval of Holoclar, a treatment involving stem cells, for use in the European Union. Holoclar is used for people with severe limbal stem cell deficiency due to burns in the eye.[8]

Research has been conducted to learn whether stem cells may be used to treat brain degeneration, such as in Parkinson's, Amyotrophic lateral sclerosis, and Alzheimer's disease.[9][10][11]

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). Pharmacological activation of endogenous neural stem cells has been reported to induce neuroprotection and behavioral recovery in adult rat models of neurological disorder.[12][13][14]

Stroke and traumatic brain injury lead to cell death, characterized by a loss of neurons and oligodendrocytes within the brain. A small clinical trial was underway in Scotland in 2013, in which stem cells were injected into the brains of stroke patients.[15]

Clinical and animal studies have been conducted into the use of stem cells in cases of spinal cord injury.[16][17][18]

The pioneering work[19] by Bodo-Eckehard Strauer has now been discredited by the identification of hundreds of factual contradictions.[20] Among several clinical trials that have reported that adult stem-cell therapy is safe and effective, powerful effects have been reported from only a few laboratories, but this has covered old[21] and recent[22] infarcts as well as heart failure not arising from myocardial infarction.[23] While initial animal studies demonstrated remarkable therapeutic effects,[24][25] later clinical trials achieved only modest, though statistically significant, improvements.[26][27] Possible reasons for this discrepancy are patient age,[28] timing of treatment[29] and the recent occurrence of a myocardial infarction.[30] It appears that these obstacles may be overcome by additional treatments which increase the effectiveness of the treatment[31] or by optimizing the methodology although these too can be controversial. Current studies vary greatly in cell-procuring techniques, cell types, cell-administration timing and procedures, and studied parameters, making it very difficult to make comparisons. Comparative studies are therefore currently needed.

Stem-cell therapy for treatment of myocardial infarction usually makes use of autologous bone-marrow stem cells (a specific type or all), however other types of adult stem cells may be used, such as adipose-derived stem cells.[32] Adult stem cell therapy for treating heart disease was commercially available in at least five continents as of 2007.[citation needed]

Possible mechanisms of recovery include:[9]

It may be possible to have adult bone-marrow cells differentiate into heart muscle cells.[9]

The first successful integration of human embryonic stem cell derived cardiomyocytes in guinea pigs (mouse hearts beat too fast) was reported in August 2012. The contraction strength was measured four weeks after the guinea pigs underwent simulated heart attacks and cell treatment. The cells contracted synchronously with the existing cells, but it is unknown if the positive results were produced mainly from paracrine as opposed to direct electromechanical effects from the human cells. Future work will focus on how to get the cells to engraft more strongly around the scar tissue. Whether treatments from embryonic or adult bone marrow stem cells will prove more effective remains to be seen.[33]

In 2013 the pioneering reports of powerful beneficial effects of autologous bone marrow stem cells on ventricular function were found to contain "hundreds" of discrepancies.[34] Critics report that of 48 reports there seemed to be just five underlying trials, and that in many cases whether they were randomized or merely observational accepter-versus-rejecter, was contradictory between reports of the same trial. One pair of reports of identical baseline characteristics and final results, was presented in two publications as, respectively, a 578 patient randomized trial and as a 391 patient observational study. Other reports required (impossible) negative standard deviations in subsets of patients, or contained fractional patients, negative NYHA classes. Overall there were many more patients published as having receiving stem cells in trials, than the number of stem cells processed in the hospital's laboratory during that time. A university investigation, closed in 2012 without reporting, was reopened in July 2013.[35]

One of the most promising benefits of stem cell therapy is the potential for cardiac tissue regeneration to reverse the tissue loss underlying the development of heart failure after cardiac injury.[36]

Initially, the observed improvements were attributed to a transdifferentiation of BM-MSCs into cardiomyocyte-like cells.[24] Given the apparent inadequacy of unmodified stem cells for heart tissue regeneration, a more promising modern technique involves treating these cells to create cardiac progenitor cells before implantation to the injured area.[37]

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 diagnosed and classified via a subspecialty of pathology known as 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.[38] Further research into this technique should have potential benefits to gene therapy, blood transfusion, and topical medicine.

Hair follicles also contain stem cells, and some researchers predict these follicle stem cells may lead to successes in treating baldness through activation of progenitor stem cells. This treatment is expected to work by activating already existing stem cells on 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.

In 2004, scientists at King's College London discovered a way to cultivate a complete tooth in mice[39] and were able to grow bioengineered teeth stand-alone in the laboratory. Researchers are confident that the tooth regeneration 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 turning into a tooth bud which, when implanted in the gums, will give rise to a new tooth, and would be expected to be grown in a time over three weeks.[40] 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.[41][42]

Research is ongoing in different fields, alligators which are polyphyodonts grow up to 50 times a successional tooth (a small replacement tooth) under each mature functional tooth for replacement once a year.[43]

Heller has reported success in re-growing cochlea hair cells with the use of embryonic stem cells.[44]

Since 2003, researchers have successfully transplanted corneal stem cells into damaged eyes to restore vision. "Sheets of retinal cells used by the team are harvested from aborted fetuses, which some people find objectionable." When these sheets are transplanted over the damaged cornea, the stem cells stimulate renewed repair, eventually restore vision.[45] 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 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.[46]

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.[47]

In 2014, researchers demonstrated that stem cells collected as biopsies from donor human corneas can prevent scar formation without provoking a rejection response in mice with corneal damage.[48]

In January 2012, The Lancet published a paper by Steven Schwartz, at UCLA's Jules Stein Eye Institute, reporting two women who had gone legally blind from macular degeneration had dramatic improvements in their vision after retinal injections of human embryonic stem cells.[49]

In June 2015, the Stem Cell Ophthalmology Treatment Study (SCOTS), the largest adult stem cell study in ophthalmology ( http://www.clinicaltrials.gov NCT # 01920867) published initial results on a patient with optic nerve disease who improved from 20/2000 to 20/40 following treatment with bone marrow derived stem cells.[50]

Diabetes patients lose the function of insulin-producing beta cells within the pancreas.[51] In recent experiments, scientists have been able to coax embryonic stem cell to turn into beta cells in the lab. In theory if the beta cell is transplanted successfully, they will be able to replace malfunctioning ones in a diabetic patient.[52]

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:[9]

Clinical case reports in the treatment 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.[53][54] 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.[55]

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.[56]

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.[57] 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.[57] Researchers are still investigating different aspects of the "soil" tissue that are conducive to regeneration.[57]

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.[58]

Human embryonic stem cells have been stimulated to form Spermatozoon-like cells, yet still slightly damaged or malformed.[59] It could potentially treat azoospermia.

In 2012, oogonial stem cells were isolated from adult mouse and human ovaries and demonstrated to be capable of forming mature oocytes.[60] These cells have the potential to treat infertility.

Destruction of the immune system by the HIV is driven by the loss of CD4+ T cells in the peripheral blood and lymphoid tissues. Viral entry into CD4+ cells is mediated by the interaction with a cellular chemokine receptor, the most common of which are CCR5 and CXCR4.1 Because subsequent viral replication requires cellular gene expression processes, activated CD4+ cells are the primary targets of productive HIV infection.[61] Recently scientists have been investigating an alternative approach to treating HIV-1/AIDS, based on the creation of a disease-resistant immune system through transplantation of autologous, gene-modified (HIV-1-resistant) hematopoietic stem and progenitor cells (GM-HSPC).[62]

On 23 January 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 aimed evaluate the drug GRNOPC1, embryonic stem cell-derived oligodendrocyte progenitor cells, on patients with acute spinal cord injury. The trial was discontinued in November 2011 so that the company could focus on therapies in the "current environment of capital scarcity and uncertain economic conditions".[63] In 2013 biotechnology and regenerative medicine company BioTime (NYSEMKT:BTX) acquired Geron's stem cell assets in a stock transaction, with the aim of restarting the clinical trial.[64]

Scientists have reported that MSCs when transfused immediately within few hours post thawing may show reduced function or show decreased efficacy in treating diseases as compared to those MSCs which are in log phase of cell growth(fresh), so cryopreserved MSCs should be brought back into log phase of cell growth in invitro culture before these are administered for clinical trials or experimental therapies, re-culturing of MSCs will help in recovering from the shock the cells get during freezing and thawing. Various clinical trials on MSCs have failed which used cryopreserved product immediately post thaw as compared to those clinical trials which used fresh MSCs.[65]

There is widespread 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.[104] There is other stem cell research that does not involve the destruction of a human embryo, and such research involves adult stem cells, amniotic stem cells and induced pluripotent stem cells.

Stem-cell research and treatment was practiced 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. The Western World has scrutinized China for its failed attempts to meet international documentation standards of these trials and procedures.[105]

State-funded companies based in the Shenzhen Hi-Tech Industrial Zone treat the symptoms of numerous disorders with adult stem-cell therapy. Development companies are currently focused on the treatment of neurodegenerative and cardiovascular disorders. The most radical successes of Chinese adult stem cell therapy have been in treating the brain. These therapies administer stem cells directly to the brain of patients with cerebral palsy, Alzheimer's, and brain injuries.[citation needed]

Since 2008 many centres and doctors tried a diversity of methods; in Lebanon proliferative and non-proliferative, in-vivo and in-vitro techniques were used. The regenerative medicine also took place in Jordan and Egypt.[citation needed]

Stem-cell treatment is currently being practiced at a clinical level in Mexico. An International Health Department Permit (COFEPRIS) is required. Authorized centers are found in Tijuana, Guadalajara and Cancun. Currently undergoing the approval process is Los Cabos. This permit allows the use of stem cell.[citation needed]

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.[106]

As of 2013, Thailand still considers Hematopoietic stem cell transplants as experimental. Kampon Sriwatanakul began with a clinical trial in October 2013 with 20 patients. 10 are going to receive stem-cell therapy for Type-2 diabetes and the other 10 will receive stem-cell therapy for emphysema. Chotinantakul's research is on Hematopoietic cells and their role for the hematopoietic system function in homeostasis and immune response.[107]

Today, Ukraine is permitted to perform clinical trials of stem-cell treatments (Order of the MH of Ukraine 630 "About carrying out clinical trials of stem cells", 2008) for the treatment of these pathologies: pancreatic necrosis, cirrhosis, hepatitis, burn disease, diabetes, multiple sclerosis, critical lower limb ischemia. The first medical institution granted the right to conduct clinical trials became the "Institute of Cell Therapy"(Kiev).

Other countries where doctors did stem cells research, trials, manipulation, storage, therapy: Brazil, Cyprus, Germany, Italy, Israel, Japan, Pakistan, Philippines, Russia, Switzerland, Turkey, United Kingdom, India and many others.

See the rest here:
Stem-cell therapy - Wikipedia, the free encyclopedia

Derivation of Ethnically Diverse Human Induced Pluripotent …

In vitro culture of primary human fibroblasts and lentivirus reprogramming

Human fibroblasts for iPSCs derivation were obtained from Coriell Institute (Camden, NJ) and reprogrammed using a single polycistronic vector using four-factor 2A (4F2A) doxycycline (DOX)-inducible lentivirus encoding mouse cDNAs for Oct4, Sox2, Klf4, and c-Myc separated by three different 2A peptides (P2A, T2A, and E2A, respectively). The lentiviral plasmids are p20321 (TetO-FUW-OSKM) and p20342 (FUW-M2rtTA) (Addgene, Cambridge, MA) originally developed by Carey et al.9. Lentiviral particles (4F2A and M2rtTA) were packaged in HEK 293T cells. The primary fibroblast cells were co-transfected using the lentivirus construct, psPAX and pCMV-VSVG vectors by calcium phosphate co-precipitation. Viral supernatants from cultures packaging each of the two viruses were pooled, filtered through a 0.45 m filter and concentrated by ultracentrifugation and stored at 80C.

The 5 human fibroblast lines were transduced by viral particles in xenofree human fibroblast culture medium10 in the presence of polybrene (8g/mL). Forty-eight hours after infection, less than 15% of fibroblasts tested immunopositive for viral-derived OCT4. The procedure was carried out in 1 well of a 6-well plate with cells at 70% confluence to allow for cell growth after viral infection an appearance of stem cell colonies. The medium was replaced two days after infection, and then daily, with xenofree hES medium plus doxycycline (1g/ml) formulated to maintain stem cell pluripotency10,11,12. After 35 days of culture, small cell clumps distinguishable from the fibroblast morphology appeared. Those that formed cell colonies with hESC-like morphology were mechanically isolated and passed on to mitotically inactivated xenofree human foreskin feeder cells (ATCC PCS-201010). Overall reprogramming efficiency by this method was calculated to be 0.002 ~ 0.004%. The iPSC colonies were expanded for several passages under xenofree conditions without doxycycline and evaluated for expression of markers of pluripotency by quantitative RT-PCR (qRT-PCR) and immunocytology.

Quantitative PCR analysis was done by isolation of total RNA from the hESC or iPSC lines and parental fibroblast lines and purification using the NucleoSpin RNA XS Total RNA isolation kit (Clontech). Reverse transcription (RT) was performed in a 20ul reaction volume using Superscript II (Invitrogen) and the cDNA reaction was diluted to a 300ul working stock volume. Primers for use in qPCR were first validated by maximally amplifying cDNA from a range of samples to confirm that a single PCR reaction product was produced and that the amplicon was of the predicted length. For validation, 10ul of cDNA from H9 hESCs (WA09, Wicell, Madison, WI), control fibroblasts (line A-2), and two of iPSC lines (A-2.2.1 & A-2.2.2) for each primer set was amplified for 36 cycles (95C 30s, 55C 30s, 72C 30s). For endogenous and transgene expression, 5ul of cDNA from each iPSC lines for each primer set was amplified for 32 cycles and resolved on a 3% nusieve agarose gel and visualized by ethidium bromide staining. Quantitative PCRs contained 10ng of cDNA, 400nM of each primer, and SYBR Green PCR Master Mix (AppliedBiosystems). Each sample was analyzed by triplicate by an ABI PRISM 7000 sequence detection system. Data was analyzed using the systems software. The expression of gene of interest was normalized to GAPDH in all cases and compared with hESCs.

We used the MycoAlertTM PLUS Assay mycoplasma detection kit (Lonza, Allendale, NJ) essentially as manufacturers instructions. Briefly, after centrifugation (1500rpm, 5min) of cell supernatant during passage of suspension iPSC cultures, the supernatants were transferred into luminescence compatible tubes (Corning Inc., Corning, NJ). The viable mycoplasma was lysed to allow enzymes to react with MycoAlertTM PLUS substrate, catalyzing the conversion of ADP to ATP. The level of ATP in the sample both before (reading A; ATP background) and after (reading B) the addition of MycoAlertTM PLUS substrate was assessed using a luminometer (Victor3, Perkin-Elmer, Waltham, Massachusetts, USA), so that a ratio B/A was obtained. Reading B assesses the conversion of ADP to ATP and is a monitor of contaminated samples. If the ratio of B/A is greater than 1 the cell culture was considered to be contaminated by mycoplasma. For control samples, the MycoAlert TM assay positive and negative control set was used.

Ethnically diverse-induced pluripotent stem cell (ED-iPSC) lines maintained on human foreskin fibroblast feeders were transferred to feeder-free conditions in non-tissue culture treated dishes coated with xenofree vitronectin (StemCell Technologies, Vancouver, Canada) or 1:100 Matrigel (10mg/ml; BD Biosciences, San Jose, CA) diluted into Hanks Buffered Saline Solution (Gibco HBSS; Life Technologies, Grand Island, NY). Cells were maintained in mTeSR2 complete media (StemCell Technologies, Vancouver, Canada) and mechanically passaged between days 5 and 7. Media was replaced on day 1 after the first passage of the series and cells grown overnight. On day 2, slow release Stem Beads FGF2 (20 microliters of PLGA beads loaded with hFGF2; StemBeads; Stem Culture Inc., Rensselaer, NY) were added with fresh mTeSR2 media. Media changes were done every 3 days with Stem Beads FGF2 and mTeSR2. Preparation of uniform sized EBs from iPSCs colonies was done in custom lithography template microarrays (LTA) generated in-house. Chemical dissociation of the stem cell colonies into single cell suspension was done before and loading of the cells into LTA- polydimethylsiloxane (PDMS) grids in mTeSR2 media in the presence of 10M Rock inhibitor (Sigma-Aldrich, St. Louis, MO) at day 0. Stem cells were maintained in grids for five days with media changes every two days. For directed multi-lineage early differentiation we used the Human Pluripotent Stem Cell Functional Identification Kit (R&D Systems, Minneapolis, MN).

For immunocytology of biomarkers in iPSC colonies, cells were prepared by two methods. Cells were fixed using 4% paraformaldehyde in PBS for 15min at room temperature and blocked by incubating cells for 90min in a solution containing 3% normal donkey serum and permeabilized by 0.1% Triton-X 100 for 10min before antibody addition. Incubations with the primary antibodies of anti-Nanog (Santa Cruz Biotechnology, Dallas, TX) and anti-SSEA4 (Santa Cruz Biotechnology, Dallas, TX) were done at 4C overnight, followed by incubation with a secondary antibody conjugated with Alexa 647 or Alexa 488 (Abcam, Cambridge, MA). After rinsing with phosphate buffered saline (PBS), the DNA was stained with bisBenzimide H 33258 (Sigma-Aldrich, St. Louis, MO) and cells imaged using a digital camera connected to a Nikon TE-2000 inverted microscope.

Phase imaging for in vitro differentiated samples was done on a Nikon 80i epifluorescence microscope using a PLAN 100.30 NA DL objective and images captured with a cooled QICam CCD camera. Fluorescent images were obtained on a Leica SP5 Laser Scanning Confocal Microscope using either HC PL FLUOTAR 100.30 NA or HCX PL APO CS 20X .70 NA objectives and also on a Zeiss AxioObserver Z1 Inverted Microscope with Colibri LED illumination, using a 100X oil 1.45 NA PlanFLUAR or 63X Plan-Apochromat 1.4 NA oil DIC objectives. Images were captured with a Hamamatsu ORCA ER CCD camera and Zeiss Axiovision Rel 4.8 acquisition software. Figures were compiled using Adobe Photoshop (Adobe Systems Inc., San Jose, CA) and Microsoft PowerPoint (Microsoft Corp., Redmond, WA) software.

The immunocytology of 2D cell cultures or three dimensional EBs was done by first fixing cells for 10 minutes at room temperature in 4% paraformaldehyde and stored overnight in PBS+0.1% Tween20 at 4C. Immediately before incubation with antibodies, the cells were permeabilized with PBS+0.5% Triton X-100 for 1 hour at 4C. Nonspecific binding was blocked by 20 minute incubation in 1% BSA in HBSS and followed by a single HBSS wash. Antibodies used for gauging pluripotency recognized Oct4A C-10 (Santa Cruz Biotechnology, Dallas, TX) and anti-SSEA4 (Millipore, Billerica, MA) (1:1000 each). Analysis of lineage commitment to differentiation was done using antibodies to OTX2 (ectoderm), SOX17 (endoderm), and Brachyury (mesoderm; 1:100 each) provided in the Human Pluripotent Stem Cell Functional Identification Kit (R&D Systems, Minneapolis, MN). Secondary antibodies were either AlexaFluor 488 or AlexaFluor 594 (A-11001, A-11037, Invitrogen, Carlsbad, CA). Nuclei were stained with bisBenzimide H 33258 (Sigma-Aldrich, St. Louis, MO) at 4C overnight and followed by washing one hour in HBSS at 4C. Samples were mounted in ProLong Gold antifade reagent (Life Technologies, Grand Island, NY) at 20C overnight in the dark before imaging immediately or storing at 4C.

Approximately 2 million ED-iPSCs were injected subcutaneously in the flank region of NOD scid gamma (NSG) mice (The Jackson Lab, Bar harbor, ME). After 1224 weeks, teratomas were formed from 10 iPSC lines, and tumors were excised & fixed in 10% normal buffered formalin (NBF) overnight. The samples were processed for histology by the Division of Human Pathology at MSU. Hematoxylin- and eosin (H&E)-stained sections were examined under a microscope.

Follow this link:
Derivation of Ethnically Diverse Human Induced Pluripotent ...

Gene Therapy and Cell Therapy Defined | ASGCT – American …

Gene therapy and cell therapy are overlapping fields of biomedical research with the goals of repairing the direct cause of genetic diseases in the DNA or cellular population, respectively. These powerful strategies are also being focused on modulating specific genes and cell subpopulations in acquired diseases in order to reestablish the normal equilibrium. In many diseases, gene and cell therapy are combined in the development of promising therapies.

In addition, these two fields have helped provide reagents, concepts, and techniques that are elucidating the finer points of gene regulation, stem cell lineage, cell-cell interactions, feedback loops, amplification loops, regenerative capacity, and remodeling.

Gene therapy is defined as a set of strategies that modify the expression of an individuals genes or that correct abnormal genes. Each strategy involves the administration of a specific DNA (or RNA).

Cell therapy is defined as the administration of live whole cells or maturation of a specific cell population in a patient for the treatment of a disease.

Gene therapy: Historically, the discovery of recombinant DNA technology in the 1970s provided the tools to efficiently develop gene therapy. Scientists used these techniques to readily manipulate viral genomes, isolate genes, identify mutations involved in human diseases, characterize and regulate gene expression, and engineer various viral vectors and non-viral vectors. Many vectors, regulatory elements, and means of transfer into animals have been tried. Taken together, the data show that each vector and set of regulatory elements provides specific expression levels and duration of expression. They exhibit an inherent tendency to bind and enter specific types of cells as well as spread into adjacent cells. The effect of the vectors and regulatory elements are able to be reproduced on adjacent genes. The effect also has a predictable survival length in the host. Although the route of administration modulates the immune response to the vector, each vector has a relatively inherent ability, whether low, medium or high, to induce an immune response to the transduced cells and the new gene products.

The development of suitable gene therapy treatments for many genetic diseases and some acquired diseases has encountered many challenges and uncovered new insights into gene interactions and regulation. Further development often involves uncovering basic scientific knowledge of the affected tissues, cells, and genes, as well as redesigning vectors, formulations, and regulatory cassettes for the genes.

While effective long-term treatments for anemias, hemophilia, cystic fibrosis, muscular dystrophy, Gauschers disease, lysosomal storage diseases, cardiovascular diseases, diabetes, and diseases of the bones and joints are elusive today, some success is being observed in the treatment of several types of immunodeficiency diseases, cancer, and eye disorders. Further details on the status of development of gene therapy for specific diseases are summarized here.

Cell therapy: Historically, blood transfusions were the first type of cell therapy and are now considered routine. Bone marrow transplantation has also become a well-established protocol. Bone marrow transplantation is the treatment of choice for many kinds of blood disorders, including anemias, leukemias, lymphomas, and rare immunodeficiency diseases. The key to successful bone marrow transplantation is the identification of a good "immunologically matched" donor, who is usually a close relative, such as a sibling. After finding a good match between the donors and recipients cells, the bone marrow cells of the patient (recipient) are destroyed by chemotherapy or radiation to provide room in the bone marrow for the new cells to reside. After the bone marrow cells from the matched donor are infused, the self-renewing stem cells find their way to the bone marrow and begin to replicate. They also begin to produce cells that mature into the various types of blood cells. Normal numbers of donor-derived blood cells usually appear in the circulation of the patient within a few weeks. Unfortunately, not all patients have a good immunological matched donor. Furthermore, bone marrow grafts may fail to fully repopulate the bone marrow in as many as one third of patients, and the destruction of the host bone marrow can be lethal, particularly in very ill patients. These requirements and risks restrict the utility of bone marrow transplantation to some patients.

Cell therapy is expanding its repertoire of cell types for administration. Cell therapy treatment strategies include isolation and transfer of specific stem cell populations, administration of effector cells, induction of mature cells to become pluripotent cells, and reprogramming of mature cells. Administration of large numbers of effector cells has benefited cancer patients, transplant patients with unresolved infections, and patients with chemically destroyed stem cells in the eye. For example, a few transplant patients cant resolve adenovirus and cytomegalovirus infections. A recent phase I trial administered a large number of T cells that could kill virally-infected cells to these patients. Many of these patients resolved their infections and retained immunity against these viruses. As a second example, chemical exposure can damage or cause atrophy of the limbal epithelial stem cells of the eye. Their death causes pain, light sensitivity, and cloudy vision. Transplantation of limbal epithelial stem cells for treatment of this deficiency is the first cell therapy for ocular diseases in clinical practice.

Several diseases benefit most from treatments that combine the technologies of gene and cell therapy. For example, some patients have a severe combined immunodeficiency disease (SCID) but unfortunately, do not have a suitable donor of bone marrow. Scientists have identified that patients with SCID are deficient in adenosine deaminase gene (ADA-SCID), or the common gamma chain located on the X chromosome (X-linked SCID). Several dozen patients have been treated with a combined gene and cell therapy approach. Each individuals hematopoietic stem cells were treated with a viral vector that expressed a copy of the relevant normal gene. After selection and expansion, these corrected stem cells were returned to the patients. Many patients improved and required less exogenous enzymes. However, some serious adverse events did occur and their incidence is prompting development of theoretically safer vectors and protocols. The combined approach also is pursued in several cancer therapies.

Further information on the progress and status of gene therapy and cell therapy on various diseases is listed here.

Here is the original post:
Gene Therapy and Cell Therapy Defined | ASGCT - American ...

Complete 2015-16 Induced Pluripotent Stem Cell Industry …

LONDON, Oct. 19, 2015 /PRNewswire/ -- Overview Summary

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, 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 you to get informed about rapidly-evolving market conditions and identify key industry trends that will give you an advantage over your competition?

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:

Commercializing iPSC products, technologies, and therapies Making intelligent investment decisions Launching high-demand products Selling effectively to your client base Increasing revenue Taking market share from your competition

Executive Summary

Stem cell research and experimentation have been in process for well over five decades, as stem cells have the unique ability to divide and replicate repeatedly. In addition, their "unspecialized" nature allows them to differentiate into a wide variety of specialized cell types. The possibilities arising from these characteristics have resulted in great commercial interest, with potential applications ranging from the use of stem cells in reversal and treatment of disease, to targeted cell therapy, tissue regeneration, pharmacological testing on cell-specific tissues, and more. Conditions such as Huntington's disease, Parkinson's disease, and spinal cord injuries are examples of clinical applications in which stem cells could offer benefits in halting or even reversing damage.

Traditionally, scientists have worked with both embryonic and adult stem cells for research tools, as well as for cellular therapy. While the appeal of embryonic cells has been their ability to differentiate into any type of cell, there has been significant ethical, moral, and spiritual controversy surrounding their use. Although some adult stem cells do have differentiation capacity, it is often limited in nature, which results in fewer options for use. Thus, induced pluripotent stem cells represent a promising combination of adult and embryonic stem cell characteristics.

Key Report Findings

Induced pluripotent stem cells represent one of the most promising research advances within the past decade, making this a valuable report for both executives and investors to use 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.

Key report findings include:

-Metrics, Timelines, Tables, and Graphs for the iPSC Industry -Trend Rate Data for iPSC Grants, Clinical Trials, and Scientific Publications -Analysis of iPSC Patent Environment, including Key Patents and Patent Trends -Market Segmentation -5-Year Market Size Projections (2015-2019) -Market Size Estimations, by Market Segment -Updates on Crucial iPSC Industry and Technology Trends -Analysis of iPSC Market Leaders, by Market Segment -Geographical Assessment of iPSC Innovation -SWOT Analysis for the iPSC Sector (Strengths, Weaknesses, Opportunities, Threats) -Preferred Species for iPSC Research -Influential Language for Selling to iPSC Scientists -Breakdown of the Marketing Methods, including Exposure and Response Rates -And Much More -End-User Survey of iPSC Scientists

A distinctive feature of this report is an end-user survey of 273 researchers (131 U.S. / 143 International) that identify as having induced pluripotent stem cells as a research focus. These survey findings reveal iPSC researcher needs, technical preferences, key factors influencing buying decisions, and more.

The findings can be used to make effective product development decisions, create targeted marketing messages, and produce higher prospect-to-client conversion rates. Download the full report: https://www.reportbuyer.com/product/3321312/

About Reportbuyer Reportbuyer is a leading industry intelligence solution that provides all market research reports from top publishers http://www.reportbuyer.com

For more information: Sarah Smith Research Advisor at Reportbuyer.com Email: query@reportbuyer.com Tel: +44 208 816 85 48 Website: http://www.reportbuyer.com

SOURCE ReportBuyer

RELATED LINKS http://www.reportbuyer.com

See the article here:
Complete 2015-16 Induced Pluripotent Stem Cell Industry ...

COMPLETE 2015-16 INDUCED PLURIPOTENT STEM CELL INDUSTRY REPORT

Overview Summary

Recent months have seen the first iPSC clinical trial in humans, creation of the worlds largest iPSC biobank, major funding awards, a historic challenge to the Yamanaka Patent, a Supreme Court ruling affecting industry patent rights, 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 you to get informed about rapidly-evolving market conditions and identify key industry trends that will give you an advantage over your competition?

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:

Commercializing iPSC products, technologies, and therapies Making intelligent investment decisions Launching high-demand products Selling effectively to your client base Increasing revenue Taking market share from your competition

Executive Summary

Stem cell research and experimentation have been in process for well over five decades, as stem cells have the unique ability to divide and replicate repeatedly. In addition, their unspecialized nature allows them to differentiate into a wide variety of specialized cell types. The possibilities arising from these characteristics have resulted in great commercial interest, with potential applications ranging from the use of stem cells in reversal and treatment of disease, to targeted cell therapy, tissue regeneration, pharmacological testing on cell-specific tissues, and more. Conditions such as Huntingtons disease, Parkinsons disease, and spinal cord injuries are examples of clinical applications in which stem cells could offer benefits in halting or even reversing damage.

Traditionally, scientists have worked with both embryonic and adult stem cells for research tools, as well as for cellular therapy. While the appeal of embryonic cells has been their ability to differentiate into any type of cell, there has been significant ethical, moral, and spiritual controversy surrounding their use. Although some adult stem cells do have differentiation capacity, it is often limited in nature, which results in fewer options for use. Thus, induced pluripotent stem cells represent a promising combination of adult and embryonic stem cell characteristics.

Key Report Findings

Induced pluripotent stem cells represent one of the most promising research advances within the past decade, making this a valuable report for both executives and investors to use 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.

Key report findings include:

-Metrics, Timelines, Tables, and Graphs for the iPSC Industry -Trend Rate Data for iPSC Grants, Clinical Trials, and Scientific Publications -Analysis of iPSC Patent Environment, including Key Patents and Patent Trends -Market Segmentation -5-Year Market Size Projections (2015-2019) -Market Size Estimations, by Market Segment -Updates on Crucial iPSC Industry and Technology Trends -Analysis of iPSC Market Leaders, by Market Segment -Geographical Assessment of iPSC Innovation -SWOT Analysis for the iPSC Sector (Strengths, Weaknesses, Opportunities, Threats) -Preferred Species for iPSC Research -Influential Language for Selling to iPSC Scientists -Breakdown of the Marketing Methods, including Exposure and Response Rates -And Much More -End-User Survey of iPSC Scientists

A distinctive feature of this report is an end-user survey of 273 researchers (131 U.S. / 143 International) that identify as having induced pluripotent stem cells as a research focus. These survey findings reveal iPSC researcher needs, technical preferences, key factors influencing buying decisions, and more.

The findings can be used to make effective product development decisions, create targeted marketing messages, and produce higher prospect-to-client conversion rates.

APPENDIX A - Properties and Characteristics of Induced Pluripotent Stem Cells APPENDIX B - iPSC Patents Held by Cellular Dyamics International (Owned by Fujifilm Holdings) APPENDIX C - Current Clinical Trials Involving iPSCs (ClinicalTrialsgov Analysis) APPENDIX D - Full List of iPSC Clinical Trial Sponsors (ClinicalTrialsgov Analysis) APPENDIX E - List of Grants that Contain iPSC Search Terms within the Title (2006 to Present; RePORTer Tool) APPENDIX F - NIH Center for Regenerative Medicine (CRM) iPSC Stem Cell Line - Control, Reporter, & Differentiated Lines

Read this article:
COMPLETE 2015-16 INDUCED PLURIPOTENT STEM CELL INDUSTRY REPORT

Bipolar Cell Pathways in the Vertebrate Retina by Ralph …

Ralph Nelson and Victoria Connaughton

1. Introduction.

Retinal ganglion cells are typically only two synapses distant from retinal photoreceptors, yet ganglion cell responses are far more diverse than those of photoreceptors. The most direct pathway from photoreceptors to ganglion cells is through retinal bipolar cells. Thus, it is of great interest to understand how bipolar cells transform visual signals.

Werblin and Dowling (1) were among the first to investigate light-evoked responses of retinal bipolar cells. Based on these studies using penetrating microelectrodes, they proposed that retinal bipolar cells lacked impulse activity, and that they processed visual signals through integration of analogue signals, that is synaptic currents and non-spike-generating voltage-gated membrane currents.

Frank Werblin and John Dowling discovered the ON or OFF light-evoked physiology of retinal bipolar cells (1). They characterized these neurons as processors of analogue visual signals that did not use impulse generation. The work was done at Johns Hopkins University as a part of Frank Werblins doctoral dissertation under John Dowlings mentorship.

Werblin and Dowing also proposed that retinal bipolar cells come in two fundamental varieties: ON-center and OFF-center (Fig. 1). Both types displayed a surround region in their receptive field that opposed the center, similar to the classic, antagonistic center-surround organization earlier described for ganglion-cell receptive fields (2). Ganglion cell receptive field organization is further reviewed in the Webvision chapter on ganglion cells. ON-center bipolar cells are depolarized by small spot stimuli positioned in the receptive field center. OFF-center bipolar cells are hyperpolarized by the same stimuli. Both types are repolarized by light stimulation of the peripheral receptive field outside the center (Fig. 1). Bipolar cells with ON-OFF responses were not encountered (1). ON-OFF responses, excitation at both stimulus onset and offset, first occur among amacrine cells, neurons postsynaptic to bipolar cells.

The Werblin and Dowling characterization of bipolar-cell physiology has proved quite durable over many decades. The notion that bipolar cells do not spike has found exception for some bipolar types. Dark-adapted Mb1 (rod bipolar cells) of goldfish generate light-evoked calcium spikes. These spikes originate in bipolar-cell axon terminals (3, 4). Through genetic imaging techniques this finding has been extended to the axon terminals of many zebrafish bipolar-cell types. In these studies bipolar terminals were labeled transgenically with the Ca2+ reporter protein SyGCaMP2 and light-induced fluctuations in Ca2+ were followed by 2-photon photometry. Fully 65% of the terminals delivered a spiking Ca2+ signal (4). In the cb5b bipolar-cell type of ground squirrel retina Na+ action potentials are driven by light. Other bipolar types in this retina do not exhibit spiking (5). These results suggest that bipolar cells are responsible for significantly more of the encoding of visual signals than had been previously supposed, and that axon-terminal spiking is actively involved. Impulse generation in bipolar cells is further discussed in the section on Voltage-gated currents.

Figure 1. Retinal bipolar cells initiate ON and OFF pathways. Microelectrode recordings of voltage responses from mudpuppy retinal neurons reveal two sorts of retinal bipolar cells: those hyperpolarized by central illumination (OFF Bipolar Cell) and those depolarized by central illumination (ON Bipolar Cell). In each case membrane potential is restored by concomitant illumination of annular rings surrounding the center. Such responses are typically 10 mV in amplitude and lack impulse activity. The absolute response latency to the light step above is about 100 msec for these suprathreshold stimuli. The illustration is taken from Werblin and Dowling, 1969 (1).

Morphology and connectivity

Anatomical investigations of bipolar cells reveal a multiplicity (4-22 depending on species) of different morphological types (6-12), significantly more than the just two types that early physiology implied. The diversity of human retinal bipolar types is illustrated in Fig. 2. Nonetheless all of these are either ON- or OFF-types and their diversity results from other factors, such as differing connectivity with photoreceptors and differing postsynaptic targets, as evidenced in the diversity of dendritic and axon-terminal ramification patterns. Some bipolar cells are postsynaptic only to rods, others only to cones (Fig. 2), and still others receive mixed rod-cone input. Among cone-selective bipolar cells, some innervate only red, green, or blue cones, while others are diffuse, that is, not selective (13-19). Different bipolar types express different glutamate receptors at subsynaptic contacts with cones.

Bipolar cell axon terminals are either mono- or multistratified, depending on the location of axonal boutons and branches in the inner plexiform layer (IPL). Differing terminal position and branching morphology within the IPL suggests that different morphological types selectively innervate different types of amacrine and ganglion cell (Fig 2).In primate retinas, bipolar cells are described as diffuse or midget types, based on the extent of the dendritic arbor. Midgets contact only a single cone, while diffuse types contact multiple cones. Bipolar cells are also termed flat or invaginating (20) depending on the placement of dendritic tips, either on the surface of (flat), or penetrating within photoreceptor synaptic terminals to approach presynaptic ribbons (invaginating).Fig. 2 illustrates 11 morphological types of bipolar cell seen in Golgi-stained human retinas.

Figure 2. Dendritic and axonal stratification patterns of bipolar cell types in human retina. The illustration is courtesy of Helga Kolb.

Bipolar cell axon terminals are either mono- or multistratified, depending on the location of axonal boutons and branches in the inner plexiform layer (IPL). Differing terminal position and branching morphology within the IPL suggests that different morphological types selectively innervate different types of amacrine and ganglion cell (Fig 2).In primate retinas, bipolar cells are described as diffuse or midget types, based on the extent of the dendritic arbor. Midgets contact only a single cone, while diffuse types contact multiple cones. Bipolar cells are also termed flat or invaginating (20) depending on the placement of dendritic tips, either on the surface of (flat), or penetrating within photoreceptor synaptic terminals to approach presynaptic ribbons (invaginating).Fig. 2 illustrates 11 morphological types of bipolar cell seen in Golgi-stained human retinas.

2. Different glutamate receptor types for ON and OFF bipolar cells.

Light responses in bipolar cells are initiated by synapses with photoreceptors. Photoreceptors release only one neurotransmitter, glutamate (21); yet bipolar cells react to this stimulus with two different responses, ON-center (glutamate hyperpolarization) and OFF-center (glutamate depolarization). Different postsynaptic glutamate receptor proteins mediate these different membrane polarizing mechanisms. The different glutamate-gated responses are associated with the differential expression of either ionotropic (iGluR) glutamate receptors (OFF bipolar cells), metabotropic (mGluR) glutamate receptor types (ON bipolar cells) or glutamate transporters (ON bipolar cells). As a result, signal transduction at the photoreceptor-to-bipolar synapse has a range of properties. The process of splitting images into multiple components tuned to selective visual features begins with differentiation of different photoreceptor types but is then greatly elaborated at the synapses between photoreceptors and bipolar cells.

Metabotropic responses of ON bipolar cells: mGluR6, Go, TRPM1, Nyctalopin

The conductance of ON bipolar cells increases in the light, whereas OFF bipolar cell conductance decreases (22, 23). The decrease in OFF bipolar cell conductance is easily explained as a loss of excitation by glutamate, as light inhibits glutamate release from photoreceptors (24). The positive reversal potential of the ON bipolar cell light response, coupled with a conductance increase (22, 25), implies that glutamate blocks a cation-permeable channel. Originally a puzzle, this was the first evidence of what we now understand as the action of metabotropic glutamate receptors (mGluRs). These receptors do not form ion channels themselves, but act as isolated antennae on the cell surface sensing glutamate and activating intracellular pathways, ultimately affecting membrane potential through mechanisms several steps removed from the binding site for glutamate. Metabotropic receptors have been identified on the axon terminals of both photoreceptors (26) and bipolar cells (27) where they serve as autoreceptors regulating glutamate release. However, the expression of one specific mGluR in the subsynaptic membrane of ON bipolar cell dendrites, the APB receptor, is unique to retina, where it is used in the direct signal transmission pathway from photoreceptors to ON bipolar cells.

Figure 3. Metabotropic glutamate receptors in the ON pathway. The glutamate agonist 2-Amino-4-Phosphonobutric acid (APB, later termed DL-AP4) interferes with light responses and membrane physiology of ON-center bipolar cells in mudpuppy. A. APB abolishes light responses (the rectangular depolarizing events), hyperpolarizes the membrane potential, and increases the membrane resistance. The latter is measured by the amplitude of voltage responses to injected current pulses (arrow). B. 3 mM cobalt, a blocker for synaptic release of glutamate from photoreceptors, abolishes light responses in an ON bipolar cell and depolarizes the membrane potential. The membrane potential can be later restored by application of APB, which acts as a substitute for the missing photoreceptor glutamate. As APB is selective for a subset of metabotropic glutamate receptors, synaptic transmission of light responses to ON bipolar cells must rely on a metabotropic mechanism. The illustration is adapted from Slaughter and Miller, 1981 (28).

The mGluR6 receptor

Slaughter and Miller (28) were the first to observe that the metabotropic glutamate agonist 2-amino-4-phosphonobutric acid (APB or DL-AP4, with the L enantiomer being effective) completely blocks the light responses of ON bipolar cells. In these neurons, APB acts as a substitute for photoreceptor-released glutamate (Fig. 3AB). Thus, ON bipolar cells utilize a metabotropic pathway to sense light-induced variations in release of photoreceptor glutamate. The metabotropic receptor has been identified as mGluR6 (29, 30). Transgenic knockout mice lacking the mGluR6 gene lack the electroretinographic b-wave (Fig. 4AB), an evoked-potential component associated with ON bipolar activity (31). The relation of electroretinogram components to cellular electrophysiology is further discussed in the Webvision chapter The Electroretinogram: ERG. Immunocytochemical localization for mGluR6 shows staining in the invaginating dendritic tips of monkey bipolar cells (Fig. 5) (32). Invaginating bipolar cells are thought to be mainly ON types in primate retina. Some foveal flat contacts also stained for mGluR6 (32).

Figure 4. MGluR6 is the metabotropic glutamate receptor expressed by ON bipolar cells. Light evoked ERG responses from the eye of a wild type (A, +/+) and a mutant (B, -/-) mouse deficient in the gene encoding mGluR6. The b-wave, which originates from the light responses of ON bipolar cells is absent in the mutant mouse. The illustration is adapted from Masu et al, 1995 (31).

Figure 5. Immunostaining for the metabotropic glutamate receptor mGluR6 selectively labels dendritic tips of invaginating bipolar cells (ib) in monkey retina. The adjacent dendritic contacts from horizontal cells (h) and the flat contact (f) from a flat cone bipolar cell are not labeled, nor is the presynaptic cone pedicle (c).The illustration is from Vardi et al., 1998 (33).

The G-protein Go

In addition to mGluR6, the G-protein Go is cytoplasmically localized in the dendritic tips of ON bipolar cells (Fig. 6) (33). Removal of the alpha subunit (Go) by knockout results in b-wave loss (34), similar to the mGluR6 knockout. Go was originally localized in rod bipolar cells, known to be ON-type, in a screen of potential G-protein second messengers for the metabotropic light response (35). This suggests that Go is directly involved in the intracellular pathway following mGluR6 activation.

The ion channel coupled to the APB receptor was originally thought to be cGMP-modulated (36).The closure of ion channels following APB binding onto mGluR6 seemed to require GTP and phosphodiesterase similar to phototransduction (36). However, the exact cascade by which this happened was less clear, as blocking phosphodiesterase (PDE) activity, or adding non-hydrolyzable cGMP analogs, did not inhibit the glutamate responses generated through APB-receptors (37). Further, it was Go that suppressed glutamate-gated current in ON bipolar cells, not transducin, the G-protein of the phototransduction cascade (37). Thus, removal of cGMP appears not to be required for channel closure (37).

Figure 6. Immunostaining for Go, the the alpha subunit of the G-protein Go, localizes to the invaginating dendritic tips of a rod bipolar cell (left) and a cone bipolar cell (right) in cat retina. Go is required for light activation of ON bipolar cells. The illustration is from Vardi, 1998 (33).

The TRPM1 channel

In agreement with these findings, recent work suggests the ON-bipolar-cell ion channel downstream of the mGluR6 receptor is not cGMP-gated (38). Rather, this non-selective cation channel identified as a TRPM1-L channel appears to be regulated by Go (38-40) in conjunction with G (41). The activity of the TRPM1 channel requires the presence of mGluR6, as the channel, though present, can not be activated in mGluR6 knockout mice (42).

TRP channels, or transient receptor potential channels, first identified in Drosophila photoreceptors (43), are present in all animal groups, including vertebrates (44), and as many as 28 channel subtypes have been identified. The TRP superfamily includes 7 subfamilies separated into two groups: TRPC, TRPV, TRPM, TRPN, and TRPA channels form Group 1; TRPP and TRPML channels form Group 2. TRPM1-L or melastatin, a melanoma related TRP channel, belongs to Group 1, and is found in ON bipolar cells. All channels share structural similarities and are permeable to cations; however, there is great functional diversity among the different channel subtypes. TRP channels are involved in many sensory systems including vision, hearing, taste, temperature-sensitivity, and osmoregulation, and are also involved in human disease (44-48).

Figure 7. TRPM1 channel knockouts lack photoresponses in ON-bipolar cells. A. In a wild type mouse, antibody staining for TRPM1 reveals localization in bipolar cells. No antibody staining is evident in the knockout. B. ON-bipolar-cell patch recordings in wild type mice reveal inward currents in response to light stimulation. This is the normal response of an ON-type bipolar cell as these cells are excited by light. No inward currents occur in ON-bipolar-cell recordings from the knockout. The illustrations are from Koike et al, 2010 (38).

In retina, TRP channels have been identified on photoreceptors (49), amacrine cells (50, 51), and ON-type bipolar cells. ON-bipolars (Fig. 7A), specifically, are antigenic for TRPM1 channels (52, 53) or TRPM1-L (38, 39, 54). Immunocytochemical and/orin situhybridization studies have localized TRPM1 expression to the dendritic tips of ON-bipolar cells (38, 39, 52), though labeling is seen in cell bodies and axons as well (Fig. 7A). TRP channels are absent in OFF-type bipolar cells. TRPM1-L channel currents have a reversal potential ~0mV (38) similar to the reversal potential of glutamate-gated currents in these cells. TRPM1-L has been shown to co-localize with and/or be functionally coupled to mGluR6 (38, 40, 42, 52). In transfected CHO cells that express mGluR6, Go, and TRPM1-L, Koike and colleagues (38) showed that all three of these components must be present for glutamate-evoked whole-cell currents to be recorded. Cells expressing only mGluR6 and Go,or only Goand TRPM1-L, did not respond to glutamate application (38, 39). These findings suggest TRPM1 channels are downstream of the mGluR6 receptor and are necessary for glutamate-elicited responses in these cells. Further, TRPM1 -/- knockout mice (Fig. 7B) do not have light-evoked ON-bipolar-cell responses and there is no ERG b-wave (38, 39, 55). The loss of response is similar to that reported for mGluR6 -/- mice (Fig. 4) (31, 56), again suggesting that both mGluR6 and TRPM1 channels are required for ON-bipolar-cell photic responses. While all cone bipolar cells in mouse appear to use an mGluR6 synapse with cones, there is evidence that some of these cells may modulate a cation channel in addition to TRPM1-L. In the TRPM1 -/- mouse, the mGluR6 antagonist CPPG still blocks a minor APB-induced membrane current (52).

Figure 8a. The insertion of the fusion protein EYFP-nyctalopin into nob NYX -/- mice re-establishes nyctalpin expression. Expression can be localized with EYFP antibodies. A. DIC image of mouse retinal slice. OS, outer segments; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer; IPL, inner plexiform layer; GC, ganglion cell layer. B. Wild type mouse is not labeled by anti-GFP. C. EYFP-NYX rescue mouse shows fusion protein localization in bipolar-cell dendritic tips within the OPL. D. High magnification of C. E. peanut agglutinin reaction labels cone terminals. F. Overlay of D and E shows NYX expression (nyctalpin) is localized in cone terminals (yellow). Green localization is presumed in rod terminals. Scale bar in A, B and C is 50 m. The illustration is from Gregg et al, 2007 (57).

The proteoglycan nyctalopin

Nyctalopin is another protein expressed on the dendritic tips of ON-bipolar cells (Fig. 8a). It is encoded by the NYX gene. NYX is required for light- and glutamate-elicited responses in ON bipolar cells (57). Mutant nob mice (58) lack an ERG b-wave and are not responsive to focal applications of glutamate onto the bipolar cell dendritic arbor (57). In wild type mice ON bipolar cells respond with outward currents to this treatment, but in nob mice they do not (Fig. 9). The nob strain is an NYX -/- mutant (59). Generation of transgenic nob mice selectively expressing EYFP-nyctalopin fusion protein in bipolar cells completely rescued the mutant phenotype. Cellular expression was restricted to bipolar cells using a regulatory sequence for GABAc1, a GABA receptor subunit selectively produced by bipolar cells. In the EYFP-NYX line, the fusion protein expression was localized to the tips of ON-bipolar cells (Fig. 8a), the b-wave was restored, and inner retinal function was similar to controls (57).

Figure 8b. Morphology of zebrafish retinal neurons expressing nyctalopin. The transgenic strain contains an MYFP gene driven by the regulatory sequence for NYX (nyctalopin). A-D retinas from 3-day post fertilization (3 dpf) larvae show only bipolar cells, with a broad distribution of axonal filopodia within the inner plexiform layer (IPL). E-G. By 6 dpf the filopdial pattern of bipolar cell axons is restricted to the inner half of the IPL, a characteristic of ON bipolar morphology. The illustration is from Schroeter, Wong and Gregg, 2006 (60).

In zebrafish a membrane-targeted yellow fluorescent protein (MYFP) reporter strain has been generated using the upstream regulatory sequences for the NYX gene to express MYFP. This reporter marks a subset of ON-type bipolar cells with characteristic long axons and terminal boutons restricted to the inner half of the inner plexiform layer. Many of these also express the ON-bipolar marker protein kinase C (PKC) (60). This genetic reporter shows the complete morphology of the cells expressing the nyctalopin gene. This transgenic tool was used to follow embryonic refinement and development of axonal projection patterns for nyctalopin-expressing ON bipolars (60) (Fig. 8b).

Subsequent studies have reported that nyctalopin complexes with both mGluR6 and TRPM1 channels in ON-bipolar cells, serving a structural role that allows proper assembly and organization of the receptor and the channel (61). In addition, nyctalopin is able to modulate TRPM1 channels, as is mGluR6 (42, 62). Thus, glutamate binding onto mGluR6 activates a G-protein (Go and/or G) leading to the closure of TRPM1 channels. The receptor and the channel are held in close proximity by nyctalopin. Alteration or mutation of any of these components mGluR6, nyctalopin, TRPM1, and/or Go can lead to a loss of response by ON-bipolar cells. In agreement with this, individuals with congenital stationary night blindness (CSNB discussed below) display a loss of ON-bipolar cell responses as evidenced in an absent ERG b-wave, and mutations in the genes encoding mGluR6, nyctalpin, and TRPM1 are associated with at least 75% of CSNB cases (62).

Figure 9. Bipolar-cell glutamate responses in the nob (nyctalopin) knockout mouse. Patch recordings of glutamate-responses reveal outward, metobotropic glutamate, currents in both rod bipolar cells and ON-type cone bipolar cells (DBC) in control mice. No glutamate currents are recorded for these cell types in nob mice. OFF cone bipolar cells (HBC) respond with inward AMPA/kainate currents in both control and nob mice. The holding potential was -60 mV. Glutamate puffs are 100 msec from pipettes filled with 1-5mM glutamate. The illustration is from Gregg et al, 2007 (57).

Modulators and subtypes

Calcium ions are a modulator of the ON bipolar metabotropic ion channel. Calcium ions, entering through the TRPM1 ion channel (63, 64) affect channel function, either by directly down regulating the channel (63) or by activating calcium-dependent enzymes, such as CaMKII (65-67), which modulate ion channel conductance. cGMP has been shown to selectively enhance ON bipolar cell responses to dim light, and may play a modulatory role for the TRPM1 channel (68).

Metabotropic receptors for ON-center bipolar cells have sustained and transient subtypes (69). The molecular basis is not yet known. However, it appears that the sustained and transient responses of ON-center ganglion cells, such as the classic X- and Y-types (70), may have their origin, at least in part, in the type of glutamate receptor expressed on the bipolar cells which innervate them (71).

Glutamate transporter mediated responses of ON bipolar cells

Ionotropic glutamate receptors with transporter-like properties are also present on some ON-center bipolar-cell dendrites. When photoreceptor glutamate binds to these transporters, a Cl conductance forms and hyperpolarizes the cells in the dark (Fig. 10). Release from this Cl inhibition occurs in the light with the decrease in glutamate released from photoreceptors. This allows the bipolar cells to depolarize (Fig. 10). Like transporters, this glutamate-gated Cl mechanism requires [Na+]o in order to function. Thus far this mechanism has been found as a dendritic glutamate receptor only in cyprinid ON bipolar cells (72-74), though it is reported in turtle, salamander and mouse photoreceptors (75-78) and is also present in mammalian central nervous system (79). Interestingly it occurs on the axon terminals of mouse rod and cone bipolar cells, where it acts to regulate glutamate release through inhibitory feedback (78).

Figure 10. An alternate ON-bipolar synaptic mechanism is a glutamate-activated-chloride channel. Puffs of glutamate mimic photoreceptor dark release in patch recordings from bipolar cells in a zebrafish retinal slice. A. Glutamate-evoked currents are outwards for physiological ranges of membrane potential, which are positive to ECl (Cl reversal potential). They are inwards at more negative potentials. The results are consistent with an ON-center mechanism driven by changes in Cl conductance. B. The 63 mV reversal potential is consistent with a model where photoreceptor glutamate opens Cl channels. Glutamate gated Cl currents are called Iglu (73) and result from the binding of glutamate to excitatory amino acid (EAAT) transporters. The illustration is from Connaughton and Nelson, 2000 (72).

Some non-mammalian bipolar cells contain both the APB and the ionotropic (transporter-like) receptor on their dendrites, while other ON-cells express either the metabotropic or the ionotropic receptor but not both (72, 73). EAAT5 has been identified as the chloride-channel-forming glutamate transporter (80). The ionotropic mechanism is used for sustained transmission between cones and bipolar cells (81, 82), and is likely to be a fast mechanism as compared to metabotropic pathways, which involve multi-step intracellular pathways and are often relatively slow (22).

The classic Mb rod bipolar cell of fish makes synapses with both rods and cones. The rod synapse mediates a conductance increase with reversal potential positive to resting potential. The cone synapse mediates a conductance decrease with reversal potential negative to the resting potential (82). Both mechanisms provide ON-type photic responses. In retrospect it would appear that the rod synapse is metabotropic, while the cone synapse is transporter-like, two different, selectively directed post-synaptic glutamate mechanisms on the same neuron.

AMPA kainate receptor expression in ON bipolar cells

ON-center bipolar cells of mammals are immunoreactive for ionotropic AMPA receptors as well as metabotropic mGluR6 receptors (83-85). In figure 11 (right panel) immunoreactivity for GluR2/3, an ionotropic AMPA subunit, appears at an invaginating, ON-type ribbon contact in cat. Similarly in teleost retinas, ON-center bipolar cells are immunoreactive for ionotropic kainate receptors (86, 87). Particularly in mammals, no physiological role has been suggested for these conventional ionotropic receptors, usually associated with OFF bipolar cells, but also seen in ON-center bipolar cells. In giant danio Wong and Dowling find that bistratified cone bipolar cells mix ON-type and OFF-type glutamate receptor mechanisms, and utilize both transporter-like receptors and AMPA/kainate receptors in generating ON and OFF color responses respectively to different spectral stimuli (88).

Figure 11.Immunostaining for the ionotropic glutamate receptor GluR1 in bipolar cell dendrites contacting cone pedicles in cat retina. Red arrows point to flat contacts, the black arrow points to an invaginating contact, and the arrowheads point to synaptic ribbons in the cone pedicle. The illustration is from Qin and Pourcho, 1999 (261).

Figure 12. Immunostaining for ionotropic glutamate receptors in the dendritic tips of cat bipolar cells. GluR6/7 subunits are found in kainate receptors. GluR2/3 subunits are found in AMPA receptors. The red arrow (left, GluR6/7) points to a immuno-stained flat contact. The red arrow (right, GluR2/3) points to an immuno-stained invaginating contact. Letter labels are invaginating bipolar (ib), horizontal cell lateral element (h), and rod (r). The illustration is from Vardi et al., 1998 (83).

Ionotropic glutamate responses of OFF bipolar cells

Like ON bipolar cells, OFF bipolar cells express more than one type of glutamate receptor, though all are ionotropic. There are three principal types of ionotropic glutamate receptors (AMPA, kainate, and NMDA) as originally defined by agonist selectivity. Though immunocytochemical studies (84, 89, 90) and in situ hybridization (91) have identified specific NMDA receptor subunits in the outer retina, OFF bipolar cells have never been observed to utilize NMDA receptors in the generation of light responses. OFF bipolar cells selectively express either AMPA or kainate receptors (92, 93). These receptors resensitize at different rates after exposure to glutamate (Fig. 13), and as a result, emphasize different temporal characteristics of the light signal. Kainate-type glutamate receptors transfer the sustained characteristics of the visual stimulus. AMPA receptors are more selective for the transient components of the signal (92). In ground squirrel retina bipolar cells are selective for one or the other (93). The situation is interesting in so far as neurons using kainate receptors exclusively are rare in the central nervous system. Nonetheless, AMPA and kainate receptors on retinal bipolar cells are pharmacologically well-behaved. Bipolar-cell AMPA-type responses can be selectively suppressed by the lipophilic AMPA receptor antagonist GYKI 52466 (94). Conversely, bipolar-cell kainate-type responses are blocked by the desensitizing kainate receptor agonist SYM 2081 (95).

Figure 13. Different OFF bipolar cells re-sensitize at different rates after glutamate treatment. In whole cell patch recordings from ground squirrel retina, bipolar cells b3 and b2 are desensitized by an initial glutamate pulse (0). The time course of recovery is measured by responses to a second pulse after different delays. Type b3 (Fig. 16) bipolar cells utilize kainate-type glutamate receptors and require several seconds for complete recovery. Type b2 bipolar cells (Fig. 16) utilize AMPA-type glutamate receptors and recover 100 times faster. The illustration is from DeVries, 2000 (92, 93).

While all retinas contain ON and OFF bipolar cell pathways, it is easy to imagine that among these pathways natural selection might cause a divergence in the expression of dendritic glutamate receptor types depending on the visual requirements of the species. In agreement with this hypothesis, species-specific differences between ON and OFF bipolar cell dendritic glutamate responses have been found. For example, ionotropic glutamate channels with transporter-like pharmacology occur exclusively in ON type bipolar cells in fish retinas. Conversely in salamander, OFF bipolar cells utilize only AMPA receptors (96). This may also be the case in zebrafish retina where dissociated cells fail to respond to the kainate agonist SYM 2081 (86) and electroretinographic OFF responses (d-waves) are blocked by the AMPA antagonist GYKI 52466 (97). One might expect also that even within the broad classes of AMPA and kainate receptors, subforms may have evolved to fit particular visual niches. In salamander retina indeed, there are separate classes of AMPA receptors postsynaptic to rods and to cones (96, 98).

3 Bipolar-cell axons: ON and OFF lamination in the inner plexiform layer

In work performed at the National Institutes of Health in the mid 1970s (99, 100), it was noted that the ON or OFF property of cat retinal ganglion cells was related to the level of stratification of dendrites within the retinal inner plexiform layer. This led to a general scheme for ON and OFF layering illustrated in figure 14. The dendrites of OFF-center ganglion cells always arborize distal to the dendrites of ON-center ganglion cells. The zone of OFF-center dendritic arborization is called sublaminaa, while the zone of ON-center dendritic arborization is called sublaminab (Fig. 14). Within each sublamina ganglion cells make selective contacts with ON- or OFF-type bipolar cells. The pattern of ON and OFF layering of bipolar cell synaptic terminals and ganglion cell dendrites has proved to be a consistent pattern among all vertebrate retinas examined (101, 102). ON and OFF layering is particularly pronounced in retinas where ganglion cell types are predominantly monostratified. However, in more anatomically complex retinas, (i.e., turtle) with multistratified and/or diffusely stratified ganglion cell types, the ON vs. OFF layering pattern applies to monostratified cells only. The physiology of cells with processes ramifying throughout the IPL is more difficult to predict based on morphology alone (103).

Figure 14. Layering of ON and OFF bipolar cell axons in the cat inner plexiform layer (IPL). OFF ganglion cell (GC and GC) dendrites and OFF cone bipolar cell axons (OFF cb) co-stratify in sublamina a of the IPL. ON bipolar axons (ON cb) and ON ganglion cell dendrites co-stratify in sublamina b of the IPL. These are the parallel ON and OFF cone pathways that originate with bipolar-cell dendritic contacts with cones. The illustration is modified from Nelson et al, 1978 (100).

Stratification of cone bipolar cell axon terminals

Go here to see the original:
Bipolar Cell Pathways in the Vertebrate Retina by Ralph ...

California Stem Cell Report

Highlights Outrage about prices Industry euphemisms Handy demons California's stem cell direction

Halloween is just around the corner and some stem cell folks here in California are doing their best to wish away a particularly frightening specter.

Some of the rhetoric amounted to no more than whistling in the dark. Investors, researchers and business executives danced around what almost certainly appear to be extremely high treatment costs for stem cell treatments.

Those costs are the type that have stirred recent outrage among consumers and among some physicians. The controversy has emerged anew in the presidential race and last week knocked the stock market around a bit.

yesterday featured a woman with cystic fibrosis who said about a drug maker,

Inevitably meetings like the Mesa conference rarely deal directly with the tough and emotional issues that are typified by the ire expressed by that woman,Klyn Elsbury, who lives a few miles north of the Mesa meeting.Instead biotech executives retreat behind such euphemisms as reimbursement, which is a catch-all term for how do we make a profit.

Yesterday the matter of pricing did come before one panel. While this writer came in late and did not hear all the names, the general response could be called if we build it, they will come.

Many of the potential products being tested now involve unmet medical needs, and thus the demand could be extraordinarily high. In other words, if you want to live, you will have to pay our price.

It would be super transformative in the market place, one speaker said, if a company has produced the only drug that will save a persons life. Another said the system will eventually find a (pricing) model. Which is where whistling in the dark comes in. But if the industry doesnt directly face the emotional and medical concerns about predatory business actions, the industry, in all likelihood, will be hoist on its own pricing petard.

Lawmakers and regulators fueled by public outrage may well react to overly aggressive prices and begin to impose what could amount to some sort of profit rationing. After all good public health is a virtuous thing. And if prices stand in the way, something needs to be done about it. Or so the reasoning will go. Every politician needs a demon to rail against. Big Pharma and related stem cell firms could be that handy demon.

The argument in some circles maintains that prices will start out sky high and then decrease over time. But that does not mean the public and other payers will wait for decades and patiently pay $1 million per treatment.

That figure popped up this week in an item by UC Davis stem cell scientist Paul Knoepfler. He wrote on his blog, ipscell.com, about a pricing model that did, in fact, run as high as $1 million.

Knoepfler said the stem cell community needs to answer following question and soon.

Californias $3 billion stem cell agency, in particular, has an economic dog in the pricing hooha. The agency is in the midst of determining how to spend its last $800 million or so. It can decide to put that money into research that offers the likelihood of relatively affordable treatments or instead into $1 million cash cow therapies for Big Pharma.

What the agency does now will affect whether it vanishes in a few years for lack of funding or can find additional support from the state and/or private sources. If its only product after running through $6 billion (including interest) is a $1 million therapy, some might look askance at providing additional cash.

Read this article:
California Stem Cell Report

U.S. Stem Cell Clinic

US Stem Cell Clinic

We are proud to announce the opening of our new state of the art US Stem Cell clinic at Sawgrass Medical Center in Sunrise, Florida. Conveniently located near Fort Lauderdale and Miami international airports, our clinic will provide first class stem cell therapy to both local patients from the South Florida area and patients from around the world. There are several quality hotel options nearby and plenty of shopping and restaurants adjacent to our clinic at the Sawgrass Mills Mall. Our staff members are standing by ready to assist out-of-town patients with their travel plans.

US Stem Cell takes pride in our Medical Team. We exclusively offer adult autologous stem cell treatment here at US Stem Cell; we do not perform any other procedures. We remain committed to providing continuing education and training for our staff. This means that our patients can have complete confidence that they are receiving the highest caliber treatment available anywhere in the world.

VIDEO:PATIENT WEBINAR STEM CELLS 101

Read more from the original source:
U.S. Stem Cell Clinic