Page 1,706«..1020..1,7051,7061,7071,708..1,7201,730..»

2016 Scientific Program

Posted: December 4, 2016 at 2:44 pm

The 2016 Scientific Committee Sessions will be held Saturday, December 3, and Sunday, December 4. Each session will be offered twice. A question-and-answer period will occur at the end of each individual speaker presentation. Invited abstracts of these sessions will be published in the Program Book and on the flash drive containing the annual meeting abstracts.

All Scientific Program sessions will be recorded and made available through ASH On Demandafter the meeting.

Robert Brodsky, MD The Johns Hopkins University School of Medicine Baltimore, MD

Ross Levine, MD Memorial Sloan-Kettering Cancer Center New York, NY

Jump to:

(Select) Ad Hoc Scientific Committee on Epigenetics and Genomics Joint Session: Scientific Committee on Blood Disorders in Childhood and Scientific Committee on Red Cell Biology Joint Session: Scientific Committee on Hematopoiesis and Scientific Committee on Myeloid Biology Scientific Committee on Bone Marrow Failure Scientific Committee on Hematopathology and Clinical Laboratory Hematology Scientific Committee on Hemostasis Scientific Committee on Immunology and Host Defense Scientific Committee on Iron and Heme Scientific Committee on Lymphoid Neoplasia Scientific Committee on Myeloid Neoplasia Scientific Committee on Plasma Cell Neoplasia Scientific Committee on Platelets Scientific Committee on Stem Cells and Regenerative Medicine Scientific Committee on Thrombosis and Vascular Biology Scientific Committee on Transfusion Medicine Scientific Committee on Transplantation Biology and Cellular Therapies

Enhancers and Chromatin Landscapes in Development and Cancer

Dr. Majeti will focus on chromatin accessibility patterns during normal humanhematopoiesis and AML evolution from pre-leukemic HSCs with a detailed discussion ofcohesin complex mutants.

Dr. Aifantis will focus on how higher order chromosomal structure is altered in leukemia and how key regulators of this process are involved in hematopoietic function and gene expression.

Dr. Ren will present data on state-of-the-art approaches to map human genomic architecture and how this process is altered during malignant transformation.

Please click here to review this session.

AshAlizadeh,MD, PhD Stanford University Stanford,CA

BingRen,PhD University of San Diego La Jolla,CA Organization and Regulation of the Human Genome

IannisAifantis,PhD New York University New York,NY The Impact of 3D Chromosomal Topology in Acute Leukemia

RaviMajeti,MD, PhD Stanford University Stanford,CA Chromatin Accessibility Charts Human Hematopoiesis and Acute Myeloid Leukemia Evolution

Understanding and Repairing Faulty Red Blood Cells

Dr. Dean will focus on the biology of enhancers, gene regulatory elements that regulate transcription through long-range interactions with promoter regions and have highly tissue-specific functions, including in erythroid cells, as well as roles in promoting pathologic gene expression in disease states.

Dr. Lodish will present ongoing work focused on harnessing an integrative, mechanistic understanding of erythroid progenitor cell signaling pathways that control self-renewing cell divisions to envision novel therapies for anemias.

Dr. De Franceschi will describe the development of non-gene therapy strategies for clinical application, including approaches currently under clinical evaluation.

Dr. Cavazzana will present novel therapeutic approaches in an effort to cure the more prevalent inherited blood diseases worldwide. Results of ongoing clinical trials as well as of promising gene editing strategies will be summarized.

Please click here to review the session.

ColleenDelaney,MD, MSc Fred Hutchinson Cancer Research Center Seattle,WA

Alex C.Minella,MD BloodCenter of Wisconsin Milwaukee,WI

AnnDean,PhD National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health Bethesda,MD New Concepts in Genome Regulation

HarveyLodish,PhD Whitehead Institute for Biomedical Research Cambridge,MA PPARa Agonists and TGF Inhibitors Stimulate Red Blood Cell Production by Enhancing Self-Renewal of BFU-E Erythroid Progenitors

LuciaDe Franceschi,MD University of Verona Verona,Italy New Therapeutic Options: Alternates to Gene Therapy for Treating Hemoglobinopathies

MarinaCavazzana,MD, PhD Hpital Necker Enfants Malades Paris,France Gene Therapy Studies in Hemoglobinopathies: Successes and Challenges

Clonal Development of Hematopoietic Stem Cell Specification and Differentiation at Single Cell Resolution

Dr. Dick will describe clonal evolution of human hematopoiesis at single cell resolution.

Dr. Gottgens will present single cell molecular profiling experiments that reveal new aspects of blood stem cell regulation and their perturbation by leukemic factors.

Dr. Rothenberg will present a systems biology level understanding of the transcription networks that control lymphoid cell fate decisions.

Dr. Schroeder will present his work using transcription factor reporters to track myeloid lineage fate determination, and the instructive role of niche and environmental factors.

Please click here to review this session

YiZheng,PhD Cincinnati Children's Hospital Cincinnati,OH

H. LeightonGrimes,PhD Cincinnati Children's Hospital Cincinnati,OH

John E.Dick,PhD University Health Network Toronto,ON,Canada Molecular Events Defining Human Clonal Hematopoiesis at Single Cell Resolution

BertieGottgens,DPhil University of Cambridge Cambridge,United Kingdom Defining Cellular States, Differentiation Trajectories, and Regulatory Networks Through Single Cell Profiling

EllenRothenberg,PhD California Institute of Technology Pasadena,CA Transcription Factor Gene Fluorescent Reporters Track Lineage Fate in Lymphoid Commitment

TimmSchroeder,PhD ETH Zurich Basel,Switzerland Long-term Live Single Cell Quantification of Transcription Factor Dynamics

Ribosomes and Ribosomopathies

Dr. Barna will introduce the concept that not all ribosomes are created equal, and that translation by specialized ribosomes represents a separate layer of gene regulation that determines which mRNAs are effectively translated. Her work provides insights into how mutations in different ribosome proteins lead to a diverse spectrum of clinical features.*Please note that Dr. Barna will only be speaking at the Saturday session.*

Dr. Warren will provide structural insights into the mechanism by which mutations that cause Schwachman-Diamond anemia affect ribosome assembly.

Dr. Zon will describe a zebrafish model of Diamond Blackfan Anemia, and how chemical suppressor screens may lead to the discovery of novel therapeutics to ameliorate clinical aspects of the syndrome.

Please click here to review the session.

NancySpeck,PhD University of Pennsylvania Perelman School of Medicine Philadelphia,PA

MariaBarna,PhD Stanford University Stanford,CA Specialized Ribosomes: A New Frontier in Gene Regulation, Organismal Biology, & Evolution

Alan J.Warren,MD, PhD University of Cambridge Cambridge,United Kingdom Shwachman-Diamond Syndrome and the Quality Control of Ribosome Assembly

Leonard I.Zon,MD Harvard Medical School, Boston Childrens Hospital Cambridge,MA Modeling Diamond Blackfan Anemia and Developing Therapeutics

Minimal Residual Disease in Hematology: Why, When, and How?

Dr. Wood will describe the key immunophenotypic principles that underlie minimal residual disease detection by flow cytometry and illustrate their application and clinical significance to the monitoring of acute leukemia.

Dr. Valk will discuss minimal residual disease detection in acute myeloid leukemia by means of polymerase chain reaction approaches using the multitude of available molecular markers in the context of clonal hematopoiesis.

Dr. Druley will focus on current strategies for using RNA sequencing as a modality for minimal residual disease detection. As we now move into the era of single-cell transcriptomes and error-corrected sequencing, we may move beyond simple quantitation of chromosomal rearrangements to identify also allele- and transcript-specific profiles of cancer cells as a tool for diagnostics, therapy and mechanistic understanding.

Please click here to review the session.

TorstenHaferlach,MD MLL Munich Leukemia Laboratory Munich,Germany

BrentWood,MD, PhD Seattle Cancer Care Alliance Seattle,WA Multiparameter Flow Cytometry as a Powerful Tool

PeterValk,PhD Erasmus University Medical Center Rotterdam,Netherlands Molecular Minimal Residual Disease Detection in Acute Myeloid Leukemia

Todd E.Druley,MD, PhD Washington University School of Medicine in St. Louis St. Louis,MO Novel Technologies to Detect Minimal Residual Disease

Emerging Therapeutics to Alter Hemostasis and Thrombosis

Dr. Lenting will describe studies of the molecular interactions between factor VIII and von Willebrand factor. Detailed understanding of these interactions has recently been uncovered and can be used for development of improved long-acting factor VIII replacement therapies for treatment of hemophilia A.

Dr. Arruda will describe the discovery and biochemical characterization of factor IX Padua and translational studies. This form of factor IX has enhanced procoagulant activity and is being advanced into gene therapy trials for treatment of hemophilia B.

Dr. Coughlin will describe the discovery and characterization of protease-activated receptors (PARs). The work explained how the coagulation protease thrombin activates platelets and other cells and led to the development of the platelet inhibitory drug, vorapaxar. A crystal structure of a PAR-vorapaxar complex has helped to explain properties of the drug.

Please click here to review this session.

AlanMast,MD, PhD BloodCenter of Wisconsin Milwaukee,WI

Peter JLenting,PhD French Institute of Health and Medical Research (INSERM) Le Kremlin-Bictre,France Von Willebrand Factor Interaction with FVIII: Development of Long Acting FVIII Therapies

ValderArruda,MD, PhD The Children's Hospital of Philadelphia, University of Pennsylvania Philadelphia,PA Factor IX Padua: From Biochemistry to Gene Therapy

ShaunCoughlin,MD, PhD University of California San Francisco San Francisco,CA PAR1 Antagonists Development and Clinical Utility

Innate Immunity: The Green Light to Adaptive Responses

Dr. Trinchieri will discuss the role of inflammation, innate resistance and commensal microbiota in carcinogenesis, cancer progression and cancer therapy.

Dr. Gajewski will discuss innate immune sensing of cancer via the host Stimulator of INterferon Genes (STING) pathway and how this presents therapeutic opportunities to activate effective anti-tumor immunity.

Please click here to review the session.

StanleyRiddell,MD Fred Hutchinson Cancer Research Center Seattle,WA

GiorgioTrinchieri,MD National Cancer Institute, National Institutes of Health Bethesda, Innate Immune Signaling in Regulation of Immunity

ThomasGajewski,MD, PhD University of Chicago Medical Center Chicago,IL Innate Immune Sensing in Anti-Tumor Immunity and Cancer Immunotherapy

From Iron Trafficking to Iron Traffic Jam

Dr. Carlomagno will discuss recent findings on the importance of ferritinophagy to maintain iron homeostasis in vivo. She will also present new data on the role of iron in regulating cell cycle progression and genome stability

Dr. Lakhal-Littleton will discuss the studies in tissue-specific and global hepcidin/ferroportin gene knockouts;offering insight into the interplay between cellular and systemic mechanisms in the regulation of iron levels in the heart and in its healthy functioning. Her studies raise the possibility that the hepcidin/ferroportin axis may also be important in other hepcidin and ferroportin-expressing tissues such as the kidney, the brain and the placenta.

Dr. Knutson will present recent insights from studies of knockout mouse models that aim to identify how various cells and organs, including the heart, take up non-transferrin-bound iron.

Please click here to review this session.

Maria DomenicaCappellini,MD University of Milan - Fondazione iRCCS Ca' Granda Ospedale Policlinico Milan Milan,Italy

FrancescaCarlomagno,MD, PhD Federico II University of Naples Naples,Italy Ferritinophagy and Cell Cycle Control

SamiraLakhal-Littleton,DPhil University of Oxford Oxford,United Kingdom Ferroportin Mediated Control of Iron Metabolism and Disease

MitchellKnutson,PhD University of Florida Gainesville,FL Non-Transferrin-Mediated Iron Delivery

Emerging Biology Leading to New Therapies in Follicular Lymphoma

Dr. Pasqualucci will introduce general concepts about the cell of origin in follicular lymphoma and the mechanisms associated with clonal evolution. She will then examine the genetic events that take place early in the history of the tumor clone and focus on the role of histone/chromatin modifier genes, including the methyltransferase KMT2D and the acetyltransferases CREBBP/EP300, in the stepwise progression of the disease from a subclinical state to a pathological entity.

Dr. Fitzgibbon will provide an introduction to genomic discovery in follicular lymphoma. He will review the next generation sequencing tools that are being used to identify genetic predisposition factors and to perform molecular profiling to identify signaling mutations that may be targeted therapeutically and which provide insights into disease prognosis.

Dr. Smith will focus her discussion on emerging new treatment approaches for follicular lymphoma based on the novel concepts and new targets described by Drs. Nadel and Fitzgibbon. She will focus on the disease heterogeneity and prognosis, the clinical unmet needs, and how clinical integration of the new molecular tools is leading to an evolution in the therapeutic regimens for patients with follicular lymphoma.

Please click here to review this session.

WendyStock,MD The University of Chicago Chicago,IL

LauraPasqualucci,MD Columbia University New York City,NY Genetic-Driven Disruption of Epigenetic Circuits As Early Steps In The Pathogenesis Of Follicular Lymphoma

JudeFitzgibbon,PhD Queen Mary University of London London,United Kingdom Genomic Discovery, Prognosis, and Target Therapy Development

Sonali M.Smith,MD The University of Chicago Medicine Chicago,IL Follicular Lymphoma Therapy Based on Biological Insights and Novel Concepts

Focusing on Myeloid Neoplasia Through Splicing

Dr. Krainer will update the audience on the spliceosoma complex and splicing machinery. He will discuss functional implications in normal and pathological conditions.

Dr. Halene will focus on the pathogenetic mechanisms underlying specific mutations in MDS.

Dr. Walter will discuss the clinical implications of spliceosome gene mutations in MDS, their distribution in diverse subgroups and their prognostic significance. He will explore novel therapeutic approachesbased on use of drugs that modulate splicing to treat spliceosome mutant MDS.

Please click here to review this session.

CristinaMecucci,MD, PhD University of Perugia Perugia,Italy

AdrianKrainer,PhD Cold Spring Harbor Laboratory Cold Spring Harbor,NY Spliceosome: Physiology and Disease Pathogenesis

StephanieHalene,MD, PhD Yale University School of Medicine New Haven,CT Functional Consequences of Spliceosome Mutations

Matthew J.Walter,MD Washington University in St. Louis St. Louis,MO Clinical Implications of Spliceosome Mutations: Epidemiology, Clonal Hematopoiesis, and Potential Therapeutic Strategies

Visit link:
2016 Scientific Program

Posted in Pennsylvania Stem Cells | Comments Off on 2016 Scientific Program

From Stem Cells to Human Development – September 2016 …

Posted: December 4, 2016 at 2:42 pm

Organisers: Olivier Pourqui, Benoit Bruneau, Gordon Keller and Austin Smith

Date: 25th 28th September 2016

Location: Southbridge Hotel & Conference Center, Massachusetts, USA

Our understanding of human embryonic development is limited by the experimental inaccessibility of the system. Thus, we have been forced to make assumptions about how humans develop based on our knowledge of other mammals, especially the mouse. However, the recent explosion in stem cell research, particularly the generation of human pluripotent stem cells and the development of organoid culture systems, has provided new opportunities for investigating lineage choice, cell differentiation, tissue organisation and even organ morphogenesis using human cells. Such work promises not only to provide a more complete knowledge of our own developmental origins, but also to inform our efforts to understand and treat developmental disorders and, perhaps most importantly, to help bring regenerative therapies to the clinic.

Following on from our highly successful inaugural meeting in 2014, the second in this series of meetings From Stem Cells to Human Development brings together scientists with a common interest in understanding human development using stem cell systems. Topics to be discussed will include the regulation of pluripotency and differentiation, development of the major lineages and tissue morphogenesis, as well as translational aspects of human stem cell research.

The fees include;

It is expected that all attendees will stay for the duration of the Meeting.

A limited number of day delegate places are available, please email meetings@biologists.com for more information.

Southbridge Hotel & Conference Center is located in the USA, minutes from Sturbridge and less than an hours drive from Boston, Springfield, Hartford, CT, and Providence, RI. The building was originally constructed as an optical factory and has been refurnished and remodelled into a superb conference centre.

Southbridge Hotel & Conference Center 14 Mechanic Street Southbridge MA 01550 USA Tel: +1 508 765 8000

Read the rest here:
From Stem Cells to Human Development - September 2016 ...

Posted in Massachusetts Stem Cells | Comments Off on From Stem Cells to Human Development – September 2016 …

Utah Stem Cells – Sports Medicine – 7430 Creek Rd, Sandy …

Posted: December 3, 2016 at 7:46 am

Specialties

We specialize in Stem Cell Joint Pain Treatment, Bioidentical Hormones for men and women, Stem Cell Aesthetics and Medical Weight Loss.

Established in 2015.

Utah Stem Cells was founded for the purpose of developing a new and exciting concept in a medical wellness center. Utilizing the latest advancements in stem cell technology, all of our services are specifically designed to enhance the quality of your life. We focus entirely on treatments that will help you feel stronger, with pain free joints, better mood, and a more beautiful appearance. You will look great and feel even better.

Dr. Bill Cimikoski, Medical Director of Utah Stem Cells, is a Medical Toxicologist that specializes in Stem Cell Joint Regeneration-a foremost authority featured on HealthLine TV and ABC's Good 4 Utah. Assisted by experienced and trained nurses and physician assistants, Dr Bill offers the treatments that can benefit you the most, while making sure that from a toxicology perspective won't hurt in the long term.

Excerpt from:
Utah Stem Cells - Sports Medicine - 7430 Creek Rd, Sandy ...

Posted in Utah Stem Cells | Comments Off on Utah Stem Cells – Sports Medicine – 7430 Creek Rd, Sandy …

Houndstongue | Montana Weed Control Association

Posted: December 3, 2016 at 7:44 am

(Cynoglossum officinale) Common Names

Gypsy flower, rats and mice, dog bur, beggers lice

Houndstongue is a biennial forb that forms a deep tap root and basal rosette the first year. It forms a flowering stem in its second year. The rosette leaves are broad, oblong, petioled and resemble a dogs tongue in shape. Leaves are alternate, up to one foot in length and up to three inches wide. They have smooth margins and are soft and velvety to touch. In the second year, stems form and often branch at the top of the plant. Plants can grow up to four feet in height. Flowers are five petaled, reddish-purple in color and produce four triangular, rounded seeds. They typically bloom in June and July. Seeds are small brown nutlets about 1/3 inch in length that easily attach to animals, vehicles, and humans. The entire plant has soft white hairs on it. The single tap root of houndstongue is thick, black and woody. Houndstoungue reproduces from seed only and each plant can produce up to 2,000 seeds. The plant dies after its second year.

The soft white hairs covering the plant, the basal leaves that resemble a hounds tongue, and the little brown burrs that stick to everything.

Houndstongue prefers well drained, relatively sandy and gravelly soils. It can also be found in shady areas and especially under the canopy of forests and wetter grasslands. It can be found in pastures and meadows, along roadsides and in disturbed sites.

Houndstongue carries an alkaloid poison that can kill livestock through loss of production of liver cells. Animals wont normally graze on it, but if cured in hay, it will remain toxic. Sheep are more resistant to this plant than cattle and horses. Horses are especially susceptible and symptoms of houndstongue ingestion include loss of weight, diarrhea, convulsions and even coma. As with many invaders, houndstongue does have medicinal properties as well and has been used as a remedy to acne, corn callus, eczema, and as a fever remedy.

Visit our library for additional articles on Houndstongue

Photo credits:Photo Credits: Nancy Chow; Matt Lavin; Photo by Richard Old, http://www.xidservices.com

Follow this link:
Houndstongue | Montana Weed Control Association

Posted in Montana Stem Cells | Comments Off on Houndstongue | Montana Weed Control Association

stem cells – decodingscience.missouri.edu

Posted: December 1, 2016 at 11:45 pm

Scientists use placental cells in lab to study virusBy Phillip Sitter | MU Bond Life Sciences Center

Megan Sheridan, an MU grad student, removes the base solution from a demonstrated sample of stem cells that will be grown into placental cells for study of Zika virus. Within four days of exposure to the correct hormones, the stem cells express genes of placental cells, and within another day start producing placental hormones. The cells are infected with Zika at day four to ensure maximum measurable interaction, as the stem cells naturally die in culture after about ten days. | photo by Phillip Sitter, Bond LSC

Scientists believe they have a better way to study how Zika virus can spread from a pregnant mother to her fetus and their technique doesnt even involve observations of babies in the womb or post-natal examinations.

As soon as we heard about Zika, everybodys light bulbs turned on, said Megan Sheridan, a graduate student at the University of Missouri Bond Life Sciences Center.

Sheridan works in the lab of Toshihiko Ezashi at Bond LSC, and she, in turn, is part of a cross-campus team researching Zika with R. Michael Roberts, Alexander Franz, Danny Schust and Ezashi.

Roberts lab studies pluripotent stem cells progenitor cells which can develop into any other type of cell in the body.

We use the proper signals to drive stem cells to become like placental cells, Sheridan explained. With this capability to stimulate stem cells with growth hormones and inhibitors at opportune moments, Roberts researchers realized they could create enough placental cells to create an environment similar to that of a womb in very early stages of pregnancy.

Megan Sheridan sits in front of a demonstration of her work with pluripotent stem cells. Sheridan is a graduate student who works in Toshihiko Ezashis lab, where she produces cells with placental characteristics from the stem cells in order to study placenta interaction with Zika virus. | photo by Phillip Sitter, Bond LSC

This is something which Sheridan thinks hasnt been done before in regards to studying placental interaction with Zika. Their technique could give a look into the first trimester, when epidemiological studies say a fetus is most susceptible to infection.

Roberts lab is trying to understand the placental barriers vulnerability to Zika virus in its early stage of pregnancy. During this time, an infection could occur even before the mother is aware she is pregnant.

If the lab uses their technique to understand how Zika virus enters placental cells, then potentially they could also learn how to strengthen the placenta as a barrier to Zika and make it a first line of defense against infection of the fetus in the womb. If developing babies dont get infected with Zika, then they wont suffer the consequences of birth defects.

One such defect is microcephaly where a baby is born with a smaller than expected head, which may in turn be a sign that their brain has not fully developed. While infection with Zika virus is rarely fatal or otherwise severe in itself many people dont even develop symptoms birth defects like microcephaly could cause further developmental problems like delays in learning how to speak and walk, intellectual disabilities, difficulty swallowing and problems with hearing and vision, according to global health organizations.

Microcephaly only became a widely documented effect of Zika after a particular strain surged across South and Central America with the infected mosquitoes that carry it, Sheridan explained, but this may be in part because previous Zika infections and outbreaks were themselves poorly documented.

While birth defects caused by Zika have drawn much media attention as the disease has spread northward through our hemisphere from Brazil, studies focusing on infection in the womb have only used placental material that has come to term. This may not be the most accurate way to see how the placenta gets infected in the first place early in pregnancy.

The pathway of Zika virus infection in lab mice isnt really comparable to human infection, because mice arent infected with this virus naturally. Only lab mice that have had their genomes altered to be able to acquire the virus have susceptibility to the infection that can be modeled.

Roberts lab is currently working with the African strain of Zika and obtained strains from Southeast Asia and Central America recently. Theres about a 99 percent genetic similarity across strains, Sheridan said.

Zika virus was first discovered in Africa in Uganda in 1947, according to the Centers for Disease Control and Prevention. The first human case was documented in 1952, and subsequent outbreaks also occurred in Southeast Asia and the Pacific Islands. The Pan American Health Organization issued an alert about the confirmed arrival of the virus in Brazil in May 2015.

The lab has completed Zika infections of some of their stem cell-produced placental cells. Sheridan reassured that even though the lab works with live viruses, Zika is not airborne, and none of their work involves mosquitoes.

Roberts lab submitted one grant application earlier this year to the National Institutes of Health for funding for their research. While that application was denied, Sheridan said that they have a lot more preliminary data now and are hoping to submit a revised grant soon.

She said that their original work was highly scored, but the funding level is still low, meaning that obtaining funds for research into Zika virus is highly competitive nationally.

Legislation to fund more efforts into studying and preventing transmission of Zika virus is caught in congressional gridlock, according to The New York Times and other media outlets.

In the mean time, as the Roberts lab prepares its next grant application submission, Sheridan said of her efforts that she is working hard to make progress on the project as quickly as possible.

Please visit the CDCs dedicated page for more information on Zika virus including advice for travellers and pregnant women, description of symptoms and treatment, steps you can take to control mosquitoes and prevent other means of transmission of the virus and more background on the history and effects of the disease.

View post:
stem cells - decodingscience.missouri.edu

Posted in Missouri Stem Cells | Comments Off on stem cells – decodingscience.missouri.edu

Stem cell controversy – Wikipedia

Posted: December 1, 2016 at 11:44 pm

The stem cell controversy is the consideration of the ethics of research involving the development, use, and destruction of human embryos. Most commonly, this controversy focuses on embryonic stem cells. Not all stem cell research involves the human embryos. For example, adult stem cells, amniotic stem cells, and induced pluripotent stem cells do not involve creating, using, or destroying human embryos, thus are minimally, if at all, controversial. Many less controversial sources of acquiring stem cells include using cells from the umbilical cord, breast milk, and bone marrow, which are not pluripotent.

For many decades, stem cells have played an important role in medical research, beginning in 1868 when Ernst Haeckel first used the phrase to describe the fertilized egg which eventually gestates into an organism. The term was later used in 1886 by William Sedgwick to describe the parts of a plant that grow and regenerate. Further work by Alexander Maximow and Leroy Stevens introduced the concept that stem cells are pluripotent, i.e. able to become many types of different cell. This significant discovery led to the first human bone marrow transplant by E. Donnal Thomas in 1968, which although successful in saving lives, has generated much controversy since. This has included the many complications inherent in stem cell transplantation (almost 200 allogeneic marrow transplants were performed in humans, with no long-term successes before the first successful treatment was made), through to more modern problems, such as how many cells are sufficient for engraftment of various types of hematopoietic stem cell transplants, whether older patients should undergo transplant therapy, and the role of irradiation-based therapies in preparation for transplantation.

The discovery of adult stem cells led scientists to develop an interest in the role of embroynic stem cells, and in separate studies in 1981 Gail Martin and Martin Evans derived pluripotent stem cells from the embryos of mice for the first time. This paved the way for Mario Capecchi, Martin Evans, and Oliver Smithies to create the first knockout mouse, ushering in a whole new era of research on human disease.

In 1998, James Thomson and Jeffrey Jones derived the first human embryonic stem cells, with even greater potential for drug discovery and therapeutic transplantation. However, the use of the technique on human embryos led to more widespread controversy as criticism of the technique now began from the wider non-scientific public who debated the moral ethics of questions concerning research involving human embryonic cells.

Since pluripotent stem cells have the ability to differentiate into any type of cell, they are used in the development of medical treatments for a wide range of conditions. Treatments that have been proposed include treatment for physical trauma, degenerative conditions, and genetic diseases (in combination with gene therapy). Yet further treatments using stem cells could potentially be developed due to their ability to repair extensive tissue damage.[1]

Great levels of success and potential have been realized from research using adult stem cells. In early 2009, the FDA approved the first human clinical trials using embryonic stem cells. These can become any cell type of the body, excluding placental cells. This ability is called pluripotency. Only cells from an embryo at the morula stage or earlier are truly totipotent, meaning that they are able to form all cell types including placental cells. Adult stem cells are generally limited to differentiating into different cell types of their tissue of origin. However, some evidence suggests that adult stem cell plasticity may exist, increasing the number of cell types a given adult stem cell can become.

Many of the debates surrounding human embryonic stem cells concern issues such as what restrictions should be made on studies using these types of cells. At what point does one consider life to begin? Is it just to destroy an embryo cell if it has the potential to cure countless numbers of patients? Political leaders are debating how to regulate and fund research studies that involve the techniques used to remove the embryo cells. No clear consensus has emerged. Other recent discoveries may extinguish the need for embryonic stem cells.[2]

Much of the criticism has been a result of religious beliefs, and in the most high-profile case, Christian US President George W Bush signed an executive order banning the use of federal funding for any cell lines other than those already in existence, stating at the time, "My position on these issues is shaped by deeply held beliefs," and "I also believe human life is a sacred gift from our creator."[3] This ban was in part revoked by his successor Barack Obama, who stated "As a person of faith, I believe we are called to care for each other and work to ease human suffering. I believe we have been given the capacity and will to pursue this research and the humanity and conscience to do so responsibly." [4]

Some stem cell researchers are working to develop techniques of isolating stem cells that are as potent as embryonic stem cells, but do not require a human embryo.

Foremost among these was the discovery in August 2006 that adult cells can be reprogrammed into a pluripotent state by the introduction of four specific transcription factors, resulting in induced pluripotent stem cells.[5] This major breakthrough won a Nobel Prize for the discoverers, Shinya Yamanaka and John Gurdon.[6]

In an alternative technique, researchers at Harvard University, led by Kevin Eggan and Savitri Marajh, have transferred the nucleus of a somatic cell into an existing embryonic stem cell, thus creating a new stem cell line.[7]

Researchers at Advanced Cell Technology, led by Robert Lanza and Travis Wahl, reported the successful derivation of a stem cell line using a process similar to preimplantation genetic diagnosis, in which a single blastomere is extracted from a blastocyst.[8] At the 2007 meeting of the International Society for Stem Cell Research (ISSCR),[9] Lanza announced that his team had succeeded in producing three new stem cell lines without destroying the parent embryos. "These are the first human embryonic cell lines in existence that didn't result from the destruction of an embryo." Lanza is currently in discussions with the National Institutes of Health to determine whether the new technique sidesteps U.S. restrictions on federal funding for ES cell research.[10]

Anthony Atala of Wake Forest University says that the fluid surrounding the fetus has been found to contain stem cells that, when used correctly, "can be differentiated towards cell types such as fat, bone, muscle, blood vessel, nerve and liver cells". The extraction of this fluid is not thought to harm the fetus in any way. He hopes "that these cells will provide a valuable resource for tissue repair and for engineered organs, as well".[11]

The status of the human embryo and human embryonic stem cell research is a controversial issue, as with the present state of technology, the creation of a human embryonic stem cell line requires the destruction of a human embryo. Most of these embryos are discarded. Stem cell debates have motivated and reinvigorated the pro-life movement, whose members are concerned with the rights and status of the embryo as an early-aged human life. They believe that embryonic stem cell research instrumentalizes and violates the sanctity of life and is tantamount to murder.[12] The fundamental assertion of those who oppose embryonic stem cell research is the belief that human life is inviolable, combined with the belief that human life begins when a sperm cell fertilizes an egg cell to form a single cell. The view of those in favor is that these embryos would otherwise be discarded, and if used as stem cells, they can survive as a part of a living human being.

A portion of stem cell researchers use embryos that were created but not used in in vitro fertility treatments to derive new stem cell lines. Most of these embryos are to be destroyed, or stored for long periods of time, long past their viable storage life. In the United States alone, an estimated at least 400,000 such embryos exist.[13] This has led some opponents of abortion, such as Senator Orrin Hatch, to support human embryonic stem cell research.[14] See also embryo donation.

Medical researchers widely report that stem cell research has the potential to dramatically alter approaches to understanding and treating diseases, and to alleviate suffering. In the future, most medical researchers anticipate being able to use technologies derived from stem cell research to treat a variety of diseases and impairments. Spinal cord injuries and Parkinson's disease are two examples that have been championed by high-profile media personalities (for instance, Christopher Reeve and Michael J. Fox, who have lived with these conditions, respectively). The anticipated medical benefits of stem cell research add urgency to the debates, which has been appealed to by proponents of embryonic stem cell research.

In August 2000, The U.S. National Institutes of Health's Guidelines stated:

...research involving human pluripotent stem cells...promises new treatments and possible cures for many debilitating diseases and injuries, including Parkinson's disease, diabetes, heart disease, multiple sclerosis, burns and spinal cord injuries. The NIH believes the potential medical benefits of human pluripotent stem cell technology are compelling and worthy of pursuit in accordance with appropriate ethical standards.[15]

In 2006, researchers at Advanced Cell Technology of Worcester, Massachusetts, succeeded in obtaining stem cells from mouse embryos without destroying the embryos.[16] If this technique and its reliability are improved, it would alleviate some of the ethical concerns related to embryonic stem cell research.

Another technique announced in 2007 may also defuse the longstanding debate and controversy. Research teams in the United States and Japan have developed a simple and cost-effective method of reprogramming human skin cells to function much like embryonic stem cells by introducing artificial viruses. While extracting and cloning stem cells is complex and extremely expensive, the newly discovered method of reprogramming cells is much cheaper. However, the technique may disrupt the DNA in the new stem cells, resulting in damaged and cancerous tissue. More research will be required before noncancerous stem cells can be created.[17][18][19][20]

Update article to include 2009/2010 current stem cell usages in clinical trials.[21][22] The planned treatment trials will focus on the effects of oral lithium on neurological function in people with chronic spinal cord injury and those who have received umbilical cord blood mononuclear cell transplants to the spinal cord. The interest in these two treatments derives from recent reports indicating that umbilical cord blood stem cells may be beneficial for spinal cord injury and that lithium may promote regeneration and recovery of function after spinal cord injury. Both lithium and umbilical cord blood are widely available therapies that have long been used to treat diseases in humans.

This argument often goes hand-in-hand with the utilitarian argument, and can be presented in several forms:

This is usually presented as a counter-argument to using adult stem cells as an alternative that does not involve embryonic destruction.

This argument is used by opponents of embryonic destruction, as well as researchers specializing in adult stem cell research.

Pro-life supporters often claim that the use of adult stem cells from sources such as umbilical cord blood has consistently produced more promising results than the use of embryonic stem cells.[30] Furthermore, adult stem cell research may be able to make greater advances if less money and resources were channeled into embryonic stem cell research.[31]

In the past, it has been a necessity to research embryonic stem cells and in doing so destroy them for research to progress.[32] As a result of the research done with both embryonic and adult stem cells, new techniques may make the necessity for embryonic cell research obsolete. Because many of the restrictions placed on stem cell research have been based on moral dilemmas surrounding the use of embryonic cells, there will likely be rapid advancement in the field as the techniques that created those issues are becoming less of a necessity.[33] Many funding and research restrictions on embryonic cell research will not impact research on IPSCs (induced pluripotent stem cells) allowing for a promising portion of the field of research to continue relatively unhindered by the ethical issues of embryonic research.[34]

Adult stem cells have provided many different therapies for illnesses such as Parkinson's disease, leukemia, multiple sclerosis, lupus, sickle-cell anemia, and heart damage[35] (to date, embryonic stem cells have also been used in treatment),[36] Moreover, there have been many advances in adult stem cell research, including a recent study where pluripotent adult stem cells were manufactured from differentiated fibroblast by the addition of specific transcription factors.[37] Newly created stem cells were developed into an embryo and were integrated into newborn mouse tissues, analogous to the properties of embryonic stem cells.

Austria, Denmark, France, Germany, and Ireland do not allow the production of embryonic stem cell lines,[38] but the creation of embryonic stem cell lines is permitted in Finland, Greece, the Netherlands, Sweden, and the United Kingdom.[38]

In 1973, Roe v. Wade legalized abortion in the United States. Five years later, the first successful human in vitro fertilization resulted in the birth of Louise Brown in England. These developments prompted the federal government to create regulations barring the use of federal funds for research that experimented on human embryos. In 1995, the NIH Human Embryo Research Panel advised the administration of President Bill Clinton to permit federal funding for research on embryos left over from in vitro fertility treatments and also recommended federal funding of research on embryos specifically created for experimentation. In response to the panel's recommendations, the Clinton administration, citing moral and ethical concerns, declined to fund research on embryos created solely for research purposes,[39] but did agree to fund research on leftover embryos created by in vitro fertility treatments. At this point, the Congress intervened and passed the Dickey Amendment in 1995 (the final bill, which included the Dickey Amendment, was signed into law by Bill Clinton) which prohibited any federal funding for the Department of Health and Human Services be used for research that resulted in the destruction of an embryo regardless of the source of that embryo.

In 1998, privately funded research led to the breakthrough discovery of human embryonic stem cells (hESC). This prompted the Clinton administration to re-examine guidelines for federal funding of embryonic research. In 1999, the president's National Bioethics Advisory Commission recommended that hESC harvested from embryos discarded after in vitro fertility treatments, but not from embryos created expressly for experimentation, be eligible for federal funding. Though embryo destruction had been inevitable in the process of harvesting hESC in the past (this is no longer the case[40][41][42][43]), the Clinton administration had decided that it would be permissible under the Dickey Amendment to fund hESC research as long as such research did not itself directly cause the destruction of an embryo. Therefore, HHS issued its proposed regulation concerning hESC funding in 2001. Enactment of the new guidelines was delayed by the incoming George W. Bush administration which decided to reconsider the issue.

President Bush announced, on August 9, 2001, that federal funds, for the first time, would be made available for hESC research on currently existing embryonic stem cell lines. President Bush authorized research on existing human embryonic stem cell lines, not on human embryos under a specific, unrealistic timeline in which the stem cell lines must have been developed. However, the Bush Administration chose not to permit taxpayer funding for research on hESC cell lines not currently in existence, thus limiting federal funding to research in which "the life-and-death decision has already been made".[44] The Bush Administration's guidelines differ from the Clinton Administration guidelines which did not distinguish between currently existing and not-yet-existing hESC. Both the Bush and Clinton guidelines agree that the federal government should not fund hESC research that directly destroys embryos.

Neither Congress nor any administration has ever prohibited private funding of embryonic research. Public and private funding of research on adult and cord blood stem cells is unrestricted.

In April 2004, 206 members of Congress signed a letter urging President Bush to expand federal funding of embryonic stem cell research beyond what Bush had already supported.

In May 2005, the House of Representatives voted 238194 to loosen the limitations on federally funded embryonic stem-cell researchby allowing government-funded research on surplus frozen embryos from in vitro fertilization clinics to be used for stem cell research with the permission of donorsdespite Bush's promise to veto the bill if passed.[45] On July 29, 2005, Senate Majority Leader William H. Frist (R-TN), announced that he too favored loosening restrictions on federal funding of embryonic stem cell research.[46] On July 18, 2006, the Senate passed three different bills concerning stem cell research. The Senate passed the first bill (the Stem Cell Research Enhancement Act) 6337, which would have made it legal for the federal government to spend federal money on embryonic stem cell research that uses embryos left over from in vitro fertilization procedures.[47] On July 19, 2006 President Bush vetoed this bill. The second bill makes it illegal to create, grow, and abort fetuses for research purposes. The third bill would encourage research that would isolate pluripotent, i.e., embryonic-like, stem cells without the destruction of human embryos.

In 2005 and 2007, Congressman Ron Paul introduced the Cures Can Be Found Act,[48] with 10 cosponsors. With an income tax credit, the bill favors research upon nonembryonic stem cells obtained from placentas, umbilical cord blood, amniotic fluid, humans after birth, or unborn human offspring who died of natural causes; the bill was referred to committee. Paul argued that hESC research is outside of federal jurisdiction either to ban or to subsidize.[49]

Bush vetoed another bill, the Stem Cell Research Enhancement Act of 2007,[50] which would have amended the Public Health Service Act to provide for human embryonic stem cell research. The bill passed the Senate on April 11 by a vote of 63-34, then passed the House on June 7 by a vote of 247176. President Bush vetoed the bill on July 19, 2007.[51]

On March 9, 2009, President Obama removed the restriction on federal funding for newer stem cell lines. [52] Two days after Obama removed the restriction, the president then signed the Omnibus Appropriations Act of 2009, which still contained the long-standing Dickey-Wicker provision which bans federal funding of "research in which a human embryo or embryos are destroyed, discarded, or knowingly subjected to risk of injury or death;"[53] the Congressional provision effectively prevents federal funding being used to create new stem cell lines by many of the known methods. So, while scientists might not be free to create new lines with federal funding, President Obama's policy allows the potential of applying for such funding into research involving the hundreds of existing stem cell lines as well as any further lines created using private funds or state-level funding. The ability to apply for federal funding for stem cell lines created in the private sector is a significant expansion of options over the limits imposed by President Bush, who restricted funding to the 21 viable stem cell lines that were created before he announced his decision in 2001.[54] The ethical concerns raised during Clinton's time in office continue to restrict hESC research and dozens of stem cell lines have been excluded from funding, now by judgment of an administrative office rather than presidential or legislative discretion.[55]

In 2005, the NIH funded $607 million worth of stem cell research, of which $39 million was specifically used for hESC.[56]Sigrid Fry-Revere has argued that private organizations, not the federal government, should provide funding for stem-cell research, so that shifts in public opinion and government policy would not bring valuable scientific research to a grinding halt.[57]

In 2005, the State of California took out $3 billion in bond loans to fund embryonic stem cell research in that state.[58]

China has one of the most permissive human embryonic stem cell policies in the world. In the absence of a public controversy, human embryo stem cell research is supported by policies that allow the use of human embryos and therapeutic cloning.[59]

According to Rabbi Levi Yitzchak Halperin of the Institute for Science and Jewish Law in Jerusalem, embryonic stem cell research is permitted so long as it has not been implanted in the womb. Not only is it permitted, but research is encouraged, rather than wasting it.

However in order to remove all doubt [as to the permissibility of destroying it], it is preferable not to destroy the pre-embryo unless it will otherwise not be implanted in the woman who gave the eggs (either because there are many fertilized eggs, or because one of the parties refuses to go on with the procedurethe husband or wifeor for any other reason). Certainly it should not be implanted into another woman.... The best and worthiest solution is to use it for life-saving purposes, such as for the treatment of people that suffered trauma to their nervous system, etc.

Similarly, the sole Jewish majority state, Israel, permits research on embryonic stem cells.

The Catholic Church opposes human embryonic stem cell research calling it "an absolutely unacceptable act." The Church supports research that involves stem cells from adult tissues and the umbilical cord, as it "involves no harm to human beings at any state of development."[60]

The Southern Baptist Convention opposes human embryonic stem cell research on the grounds that "Bible teaches that human beings are made in the image and likeness of God (Gen. 1:27; 9:6) and protectable human life begins at fertilization."[61] However, it supports adult stem cell research as it does "not require the destruction of embryos."[61]

The United Methodist Church opposes human embryonic stem cell research, saying, "a human embryo, even at its earliest stages, commands our reverence."[62] However, it supports adult stem cell research, stating that there are "few moral questions" raised by this issue.[62]

The Assemblies of God opposes human embryonic stem cell research, saying, it "perpetuates the evil of abortion and should be prohibited."[63]

The religion of Islam favors the stance that scientific research and development in terms of stem cell research is allowed as long as it benefits society while using the least amount of harm to the subjects. "Stem cell research is one of the most controversial topics of our time period and has raised many religious and ethical questions regarding the research being done. With there being no true guidelines set forth in the Qur'an against the study of biomedical testing, Muslims have adopted any new studies as long as the studies do not contradict another teaching in the Qur'an. One of the teachings of the Qur'an states that Whosoever saves the life of one, it shall be if he saves the life of humankind (5:32), it is this teaching that makes stem cell research acceptable in the Muslim faith because of its promise of potential medical breakthrough."[64]

The First Presidency of The Church of Jesus Christ of Latter-day Saints "has not taken a position regarding the use of embryonic stem cells for research purposes. The absence of a position should not be interpreted as support for or opposition to any other statement made by Church members, whether they are for or against embryonic stem cell research.[65]

See more here:
Stem cell controversy - Wikipedia

Posted in Massachusetts Stem Cells | Comments Off on Stem cell controversy – Wikipedia

Cell therapy – Wikipedia

Posted: December 1, 2016 at 11:41 pm

Cell therapy (also called cellular therapy or cytotherapy) is therapy in which cellular material is injected into a patient;[1] this generally means intact, living cells. For example, T cells capable of fighting cancer cells via cell-mediated immunity may be injected in the course of immunotherapy.

Cell therapy originated in the nineteenth century when scientists experimented by injecting animal material in an attempt to prevent and treat illness.[2] Although such attempts produced no positive benefit, further research found in the mid twentieth century that human cells could be used to help prevent the human body rejecting transplanted organs, leading in time to successful bone marrow transplantation.[3]

Today two distinct categories of cell therapy are recognized.[1]

The first category is cell therapy in mainstream medicine. This is the subject of intense research and the basis of potential therapeutic benefit.[4] Such research can be controversial when it involves human embryonic material.

The second category is in alternative medicine, and perpetuates the practice of injecting animal materials in an attempt to cure disease. This practice, according to the American Cancer Society, is not backed by any medical evidence of effectiveness, and can have deadly consequences.[1]

Cell therapy can be defined as therapy in which cellular material is injected into a patient.[1]

There are two branches of cell therapy: one is legitimate and established, whereby human cells are transplanted from a donor to a patient; the other is dangerous alternative medicine, whereby injected animal cells are used to attempt to treat illness.[1]

The origins of cell therapy can perhaps be traced to the nineteenth century, when Charles-douard Brown-Squard (18171894) injected animal testicle extracts in an attempt to stop the effects of aging.[2] In 1931 Paul Niehans (18821971) who has been called the inventor of cell therapy attempted to cure a patient by injecting material from calf embryos.[1] Niehans claimed to have treated many people for cancer using this technique, though his claims have never been validated by research.[1]

In 1953 researchers found that laboratory animals could be helped not to reject organ transplants by pre-inoculating them with cells from donor animals; in 1968, in Minnesota, the first successful human bone marrow transplantation took place.[3]

Bone marrow transplants have been found to be effective, along with some other kinds of human cell therapy for example in treating damaged knee cartilage.[1] In recent times, cell therapy using human material has been recognized as an important field in the treatment of human disease.[4] The experimental field of Stem cell therapy has shown promise for new types of treatment.[1]

In mainstream medicine, cell therapy is supported by a distinct healthcare industry which sees strong prospects for future growth.[5][6]

In allogeneic cell therapy the donor is a different person to the recipient of the cells.[7] In pharmaceutical manufacturing, the allogenic methodology is promising because unmatched allogenic therapies can form the basis of "off the shelf" products.[8] There is research interest in attempting to develop such products to treat conditions including Crohn's disease[9] and a variety of vascular conditions.[10]

Research into human embryonic stem cells is controversial, and regulation varies from country to country, with some countries banning it outright. Nevertheless, these cells are being investigated as the basis for a number of therapeutic applications, including possible treatments for diabetes[11] and Parkinson's disease.[12]

Cell therapy is targeted at many clinical indications in multiple organs and by several modes of cell delivery. Accordingly, the specific mechanisms of action involved in the therapies are wide ranging. However, there are two main principles by which cells facilitate therapeutic action:

Neural stem cells (NSCs) are the subject of ongoing research for possible therapeutic applications, for example for treating a number of neurological disorders such as Parkinson's disease and Huntington's disease.[20]

MSCs are immunomodulatory, multipotent and fast proliferating and these unique capabilities mean they can be used for a wide range of treatments including immune-modulatory therapy, bone and cartilage regeneration, myocardium regeneration and the treatment of Hurler syndrome, a skeletal and neurological disorder.[21]

Researchers have demonstrated the use of MSCs for the treatment of osteogenesis imperfecta (OI). Horwitz et al. transplanted bone marrow (BM) cells from human leukocyte antigen (HLA)-identical siblings to patients suffering from OI. Results show that MSCs can develop into normal osteoblasts, leading to fast bone development and reduced fracture frequencies.[22] A more recent clinical trial showed that allogeneic fetal MSCs transplanted in utero in patients with severe OI can engraft and differentiate into bone in a human fetus.[23]

Besides bone and cartilage regeneration, cardiomyocyte regeneration with autologous BM MSCs has also been reported recently. Introduction of BM MSCs following myocardial infarction (MI) resulted in significant reduction of damaged regions and improvement in heart function. Clinical trials for treatment of acute MI with Prochymal by Osiris Therapeutics are underway. Also, a clinical trial revealed huge improvements in nerve conduction velocities in Hurlers Syndrome patients infused with BM MSCs from HLA-identical siblings.[24]

HSCs possess the ability to self-renew and differentiate into all types of blood cells, especially those involved in the human immune system. Thus, they can be used to treat blood and immune disorders. Since human bone marrow (BM) grafting was first published in 1957,[25] there have been significant advancements in HSCs therapy. Following that, syngeneic marrow infusion[26] and allogeneic marrow grafting[27] were performed successfully. HSCs therapy can also render its cure by reconstituting damaged blood-forming cells and restoring the immune system after high-dose chemotherapy to eliminate disease.[28]

There are three types of HSCT: syngeneic, autologous, and allogeneic transplants.[21] Syngeneic transplantations occur between identical twins. Autologous transplantations use the HSCs obtained directly from the patient and hence do not cause any complications of tissue incompatibility; whereas allogeneic transplantations involve the use of donor HSCs, either genetically related or unrelated to the recipient. To lower the risks of transplant, which include graft rejection and Graft-Versus-Host Disease (GVHD), allogeneic HSCT must satisfy compatibility at the HLA loci (i.e. genetic matching to reduce the immunogenicity of the transplant). Mismatch of HLA loci would result in treatment-related mortality and higher risk of acute GVHD.[29]

In addition to BM derived HSCs, the use of alternative sources such as umbilical cord blood (UCB) and peripheral blood stem cells (PBSCs) has been increasing. In comparison with BM derived HSCs recipients, PBSCs recipients afflicted with myeloid malignancies reported a faster engraftment and better overall survival.[30] However, this was at the expense of increased rate of GVHD.[31] Also, the use of UCB requires less stringent HLA loci matching, although the time of engraftment is longer and graft failure rate is higher.[32][33]

In alternative medicine, cell therapy is defined as the injection of non-human cellular animal material in an attempt to treat illness.[1]Quackwatch labels this as "senseless", since "cells from the organs of one species cannot replace the cells from the organs of other species" and because a number of serious adverse effects have been reported.[34]

Of this alternative, animal-based form of cell therapy, the American Cancer Society say: "Available scientific evidence does not support claims that cell therapy is effective in treating cancer or any other disease. In may in fact be lethal ...".[1]

More:
Cell therapy - Wikipedia

Posted in Cell Therapy | Comments Off on Cell therapy – Wikipedia

Idaho National Laboratory – Wikipedia

Posted: November 30, 2016 at 6:45 pm

Idaho National Laboratory Motto The energy of innovation Established 1949 Researchtype Nuclear energy, national security, energy, and environment Budget ~ $1 billion (2010) Director Mark Peters Staff ~ 4,000 (2016) Location Idaho Falls, Idaho, U.S. & a large area to the west Campus 890sqmi (2,310km2)

Operating agency

Idaho National Laboratory (INL) is an 890-square-mile (2,310km2) complex located in the high desert of eastern Idaho, between the town of Arco to the west and the cities of Idaho Falls and Blackfoot to the east. It lies within Butte, Bingham, Bonneville, and Jefferson counties. The lab currently employs approximately 4,000 people.

The federal research facility was established in 1949 as the "National Reactor Testing Station" (NRTS).[1] In 1975, the Atomic Energy Commission (AEC) was divided into the Energy Research and Development Administration (ERDA) and the Nuclear Regulatory Commission (NRC). The Idaho site was for a short time named ERDA and then subsequently renamed to the "Idaho National Engineering Laboratory" (INEL) in 1977 with the creation of the Department of Energy (DOE) under President Carter. After two decades as INEL, the name was changed again to the "Idaho National Engineering and Environmental Laboratory" (INEEL) in 1997. Throughout its lifetime, there have been more than 50 one-of-a-kind nuclear reactors built by various organizations at the facility for testing; all but three are out of service.

On Feb. 1, 2005, Battelle Energy Alliance took over operation of the lab from Bechtel, merged with Argonne National Laboratory-West, and the facility name was changed to "Idaho National Laboratory" (INL).[2] At this time the site's clean-up activities were moved to a separate contract, the Idaho Cleanup Project, which is currently managed by Fluor Idaho, LLC. Research activities were consolidated in the newly named Idaho National Laboratory.

In the Snake River Plain, most of INL is desert with scrub vegetation and a number of facilities scattered throughout the area; the average elevation of the complex is 5,000 feet (1,520m) above sea level. A few publicly accessible highways go through the vast INL, but most of the area (except EBR-I) is restricted to authorized personnel and requires appropriate security clearance. The tiny town of Atomic City is on the INL's southern border, and the Craters of the Moon National Monument is to the southwest.

One part of this program to develop improved nuclear power plants is the Next Generation Nuclear Plant or NGNP, which would be the demonstration of a new way to use nuclear energy for more than electricity. The heat generated from nuclear fission in the plant could provide process heat for hydrogen production and other industrial purposes, while also generating electricity. And the NGNP would use a high-temperature gas reactor,[3] which would have redundant safety systems that rely on natural physical processes more than human or mechanical intervention.

INL is working with private industry to design, plan and eventually build the NGNP. It was commissioned to lead this effort by the U.S. Department of Energy as a result of the Energy Policy Act of 2005.[4]

The Fuel Cycle Research & Development program aims to help expand nuclear energys benefits by addressing some of the issues inherent to the current life cycle of nuclear reactor fuel in the United States. These efforts strive to make nuclear energy's expansion safe, secure, economic and sustainable.

Currently, the United States, like many other countries, employs an open-ended" nuclear fuel cycle, whereby nuclear power plant fuel is used only once and then placed in a repository for indefinite storage. One of the primary FCRD goals is to research, develop and demonstrate ways to close the fuel cycle so fuel is reused or recycled rather than being shelved before all of its energy has been used. INL coordinates many of the FCRD's national research efforts, including:

The Light Water Reactor Sustainability Program supports national efforts to do the research and gather the information necessary to demonstrate whether it is safe and prudent to apply for extensions beyond 60 years of operating life.

The Program aims to safely and economically extend the service lives of the more than 100 electricity-generating nuclear power plants in the United States. The program brings together technical information, performs important research and organizes data to be used in license-extension applications.[6]

INLs Advanced Test Reactor is a unique research reactor located approximately 50 miles (80km) from Idaho Falls, Idaho.

The Department of Energy named Advanced Test Reactor (ATR) a National Scientific User Facility in April 2007. This designation opened the facility to use by university-led scientific research groups and gives them free access to the ATR and other unique resources at INL and partner facilities.[7] In addition to a rolling proposal solicitation with two closing dates each year, INL holds an annual "Users Week" and summer session to familiarize researchers with the user facility capabilities available to them.

DOE's Nuclear Energy University Programs provides funding for university research grants, fellowships, scholarships and infrastructure upgrades.

For example, in May 2010, the program awarded $38 million for 42 university-led R&D projects at 23 U.S. universities in 17 states. In FY 2009, the program awarded about $44 million to 71 R&D projects and more than $6 million in infrastructure grants to 30 U.S. universities and colleges in 23 states.[8] INL's Center for Advanced Energy Studies administers the program for DOE. CAES is a collaboration between INL and Idaho's three public research universities: Idaho State University, Boise State University and University of Idaho.

The Multiphysics Methods Group (MMG) is a program at Idaho National Laboratory (under the U.S. Department of Energy) begun in 2004. It uses applications based on the multiphysics and modeling framework MOOSE to simulate complex physical and chemical reactions inside nuclear reactors . The ultimate goal of the program is to use these simulation tools to enable more efficient use of nuclear fuel, resulting in lower electricity costs and less waste products.[9] The MMG is made up of 11 members, and is led by Glen Hansen. It coordinates efforts with other specialists in academia and industry.[10]

The MMG focuses on problems within nuclear reactors related to its fuel and how heat is transferred inside the reactor. "Fuel degradation" refers to how uranium pellets and the rods they are encased in (several rods bundled together is what makes a "fuel assembly") eventually wear out over time due to high heat and irradiation inside a reactor. The group states three main objectives: "The mission of the MMG is to support the INL goal to advance the U.S. nuclear energy endeavor by:[11]

The work done by the group directly supports programs such as the Light Water Reactor Sustainability Program's research into advanced nuclear fuels.

INL's National and Homeland Security division focuses on two main areas: protecting critical infrastructure such as electricity transmission lines, utilities and wireless communications networks, and preventing the proliferation of weapons of mass destruction.

For nearly a decade, INL has been conducting vulnerability assessments and developing innovative technology to increase infrastructure resiliency. With a strong emphasis on industry collaboration and partnership, INL is enhancing electric grid reliability, control systems cybersecurity and physical security systems.[12]

INL conducts advanced cyber training and oversees simulated competitive exercises for national and international customers.[13] The lab supports cyber security and control systems programs for the departments of Homeland Security, Energy and Defense. INL staff members are frequently asked to provide guidance and leadership to standards organizations, regulatory agencies and national policy committees.

In January 2011, it was reported by the New York Times that the INL was allegedly responsible for some of the initial research behind the Stuxnet virus which allegedly crippled Iran's nuclear centrifuges. The INL, which teamed up with Siemens, conducted research on the P.C.S.-7 control system to identify its vulnerabilities. According to the Times, that information would later be used by the American and Israeli governments to create the Stuxnet virus.[14]

The Times article was later disputed by other journalistsincluding Forbes blogger Jeffrey Carras being both sensational and lacking verifiable facts.[15] In March 2011, Vanity Fair Magazine's cover story on Stuxnet carried INL's official response stating, "Idaho National Laboratory was not involved in the creation of the stuxnet worm. In fact, our focus is to protect and defend control systems and critical infrastructures from cyber threats like stuxnet and we are all well recognized for these efforts. We value the relationships that we have formed within the control systems industry and in no way would risk these partnerships by divulging confidential information."[16]

Building on INL's nuclear mission and legacy in reactor design and operations, the lab's engineers are developing technology, shaping policy and leading initiatives to secure the nuclear fuel cycle and prevent the proliferation of weapons of mass destruction.[17]

Under the direction of the National Nuclear Security Administration, INL and other national laboratory scientists are leading a global initiative to secure foreign stockpiles of fresh and spent highly enriched uranium and return it to secure storage for processing .[18] Other engineers are working to convert U.S. research reactors and build new reactor fuels that replace highly enriched uranium with a safer, low-enriched uranium fuel.[19] To protect against threats from the dispersal of nuclear and radiological devices, INL researchers also examine radiological materials to understand their origin and potential uses. Others have applied their knowledge to the development of detection technologies that scan and monitor containers for nuclear materials.

The laboratory's expansive desert location, nuclear facilities and wide range of source materials provide an ideal training location for military responders, law enforcement and other civilian first responders. INL routinely supports these organizations by leading classroom training, conducting field exercises and assisting in technology assessments.

INLs Advanced Vehicle Testing Activity gathers information from more than 4000 plug-in-hybrid vehicles. These vehicles, operated by a wide swath of companies, local and state governments, advocacy groups, and others are located all across the United States, Canada and Finland. Together, theyve logged a combined 1.5 million miles worth of data that is analyzed by specialists at INL.

Dozens of other types of vehicles, like hydrogen-fueled and pure electric cars, are also tested at INL. This data will help evaluate the performance and other factors that will be critical to widespread adoption of plug-in or other alternative vehicles.[20][21]

INL researchers are partnering with farmers, agricultural equipment manufacturers and universities to optimize the logistics of an industrial-scale biofuel economy. Agricultural waste products such as wheat straw; corncobs,[22][23] stalks or leaves; or bioenergy crops such as switchgrass or miscanthus could be used to create cellulosic biofuels. INL researchers are working to determine the most economic and sustainable ways to get biofuel raw materials from fields to biorefineries.[24]

INLs robotics program researches, builds, tests and refines robots that, among other things, clean up dangerous wastes, measure radiation, scout drug-smuggling tunnels, aid search-and-rescue operations, and help protect the environment.

These robots roll, crawl, fly,[25] and go under water, even in swarms[26] that communicate with each other on the go to do their jobs.

The Biological Systems department is housed in 15 laboratories with a total of 12,000 square feet (1,100m2) at the INL Research Center in Idaho Falls. The department engages in a wide variety of biological studies, including studying bacteria and other microbes that live in extreme conditions such as the extremely high temperature pools of Yellowstone National Park.[27] These types of organisms could boost the efficiency of biofuels production. Other studies related to uncommon microbes have potential in areas such as carbon dioxide sequestration and groundwater cleanup.[28]

INL is pioneering the research and testing associated with hybrid energy systems that combine multiple energy sources for optimum carbon management and energy production. For example, a nuclear reactor could provide electricity when certain renewable resources aren't available, while also providing a carbon-free source of heat and hydrogen that could be used, for example, to make liquid transportation fuels from coal.[29]

The Integrated Waste Treatment Unit (IWTU)

Construction of a new liquid waste processing facility is nearly completed at INTEC on the INL Site. It will process approximately 900,000 gallons of liquid nuclear waste using a steam reforming process to produce a granular product suitable for disposal. The facility is the first of its kind and based on a scaled prototype. The project is a part of the Department of Energy's Idaho Cleanup Project aimed at removing waste and demolishing old nuclear facilities at the INL site.[30][31][32][33]

The Instrumentation, Control and Intelligent Systems (ICIS) Distinctive Signature supports mission-related research and development in key capability areas: safeguards and control system security, sensor technologies, intelligent automation, human systems integration, and robotics and intelligent systems. These five key areas support the INL mission to ensure the nations energy security with safe, competitive, and sustainable energy systems and unique national and homeland security.[citation needed] Through its grand challenge in resilient control systems, ICIS research is providing a holistic approach to aspects of design that have often been bolt-on, including human systems, security and modeling of complex interdependency.

INL supports science, technology, engineering and math (STEM) education in classrooms across the state. Each year, the lab invests nearly $500,000 in Idaho teachers and students. Funding goes toward scholarship programs for high school graduates, technical college students and teachers who want to integrate more hands-on science activities into their lessons. INL also provides thousands of dollars worth of classroom grants to teachers seeking to upgrade their science equipment or lab infrastructure.[34]

The lab hires more than 300 interns each summer to work alongside laboratory employees. INL is listed by Vault, the online job resource site, as one of the best places in the U.S. to get an internship[35] Internships are offered to high school, undergraduate, graduate and post-graduate students in applicable fields including science, engineering, math, chemistry, business, communication and other fields.

In addition to subcontracting more than $100 million worth of work from Idaho's small businesses,[36] INL technologies are often licensed to new or existing companies for commercialization. In the past 10 years, INL has negotiated roughly 500 technology licenses. And INL technology has spawned more than 40 start-up companies since 1995.[37]

Small businesses that contract with the lab can participate in a Department of Energy program designed to enhance their capabilities. INL has worked with a variety of small businesses in this mentoring capacity, including International Management Solutions and Portage Environmental.[38]

INLs Advanced Test Reactor is much smaller than the more common electricity-producing reactors the reactor vessel measures 12 feet (3.7m) across and 36 feet (11m) high, with the core a mere 4 feet (1.2m) tall and 50inches across, and it does not generate electricity. As a special feature, it allows scientists to simultaneously test materials in multiple unique experimental environments. Research scientists can place experiments in one of the more than 70 test positions in the reactor. Each can generate unique experimental conditions.

Some have called the reactor a virtual time machine,[39] for its ability to demonstrate the effects of several years of radiation on materials in a fraction of the time.

The ATR allows scientists to place a great variety of materials in an environment with specified intensities of radiation, temperature and pressure. Specimens are then removed to examine how the time in the reactor affected the materials. The U.S. Navy is the facility's primary user, but the ATR also produces medical isotopes that can help treat cancer patients and industrial isotopes that can be used for radiography to x-ray welds on items such as skyscrapers, bridges and ship holds.

Many ATR experiments focus on materials that could make the next generation of nuclear reactors even safer and longer lasting.[40]>

The Hot Fuel Examination Facility (HFEF) gives INL researchers and other scientists the ability to examine and test highly radioactive irradiated reactor fuel and other materials.

HFEF provides 15 state-of-the-art workstations known as hot cells. For windows, each cell has leaded glass panes layered 4 feet (1.2m) thick and separated by thin layers of oil. Remote manipulators allow users to maneuver items inside the hot cell using robotic arms. And special filtered exhaust systems[41] keep indoor and outdoor air safe. At these stations, scientists and technicians can better determine the performance of irradiated fuels and materials. Scientists can also characterize materials destined for long-term storage at the Waste Isolation Pilot Plant in New Mexico.

The New Horizons mission to Pluto, which launched in 2006, is powered by a device fueled at the INL Space and Security Power Systems Facility. The Radioisotope Thermoelectric Generator (RTG) uses nonweapons-grade plutonium to produce heat and electricity for deep space missions such as this one.

Using the RTG on the New Horizons mission is a more practical power source for the satellite than solar panels because the satellite will travel to such a great distance that energy from the sun would provide insufficient power for the craft.[42] Work on the project started in late 2004 and ended with the January 2006 successful rocket launch. The team implemented the fueling, testing and delivery of the RTG for the Pluto New Horizons mission and for the next Mars rover.[43]

INL's Fuel Conditioning Facility uses electrolysis to separate certain components from used nuclear fuel rods. Unlike traditional aqueous reprocessing techniques, which dissolve the fuel rods in acid, "pyroprocessing" melts the rods and uses electricity to separate components such as uranium and sodium out of the mix. INL is using this technique to remove the sodium metal from EBR-II fuel rods so they can be safely stored in a national repository.[citation needed]

The Transient Reactor Test Facility (TREAT) is a reactor designed specifically to test new reactor fuels and materials.

The Critical Infrastructure Test Range at INL's 890-square-mile (2,300km2) Site allows researchers to conduct resiliency exercises and experiments from conceptual design to full-scale demonstration. INL also has access to a utility-scale power grid, substations, unique real-time modeling and simulation systems, and vendor-supplied Supervisory Control and Data Acquisition (SCADA) systems for demonstration and deployment exercises.[citation needed]

In addition, INL owns and operates a communications network designed to research and test cellular, mobile and emerging Internet communication protocols and technology, with both fixed and mobile 3-G platforms that allow testing and demonstration within a range of experimental frequencies in a low-background environment.

This unique partnership between INL and Idaho's three public research universities Idaho State University, University of Idaho and Boise State University boasts a wealth of research expertise. Its researchers, who have access to each partner institutions equipment and infrastructure, have competed for and won millions of dollars in national funding for their projects. CAES possesses capabilities and infrastructure unique to the region and nation. The centers laboratories are equipped with state-of-the-art research instruments and tools, including a Local Electrode Atom Probe (LEAP) and a Computer Assisted Virtual Environment (CAVE).

The Matched Index of Refraction facility is the largest such facility in the world. Using light mineral oil, the facility allows researchers to use fused quartz models built to scale to study the flow of liquids inside and around objects with complicated geometries, such as the core of a nuclear reactor. The facility is basically a giant loop through which the mostly transparent oil is pumped at variable speeds. Special lasers perform Doppler velocimetry, that produces a 3-D image allowing inspection of an objects flow properties. Observers can also watch the flow themselves through the polycarbonate viewing panes near the laser equipment.[44]Video: Matched Index of Refraction Flow Facility

INLs geocentrifuge helps researchers, among other efforts, improve models of how liquids and contaminants move through engineered caps and barriers used in underground waste disposal facilities.[45]

The INL centrifuge is one of fewer than 25 geocentrifuges larger than two meters (about 6 feet) in the United States.[45] The centrifuge, located next to the INL Research Center in Idaho Falls, can be operated remotely by computer and is capable of applying 130 times the force of earths gravity on a sample.[46]

Many of the experiments that use the geocentrifuge require it to run for hundreds of hours in order to correctly simulate several years worth of gravitational effects. The payload is monitored by an onboard computer and can be relayed to a remote monitoring station outside the centrifuges chamber where technicians can observe developments.[46]

Much of current knowledge about how nuclear reactors behave and misbehave was discovered at what is now Idaho National Laboratory. John Grossenbacher, former INL director, said, "The history of nuclear energy for peaceful application has principally been written in Idaho."[47]

More than 50 reactors have been built by various organizations at what is commonly called the Site, including the ones that gave the world its first usable amount of electricity produced from nuclear power and the power plant for the worlds first nuclear submarine. Although many are now decommissioned, these facilities represent the largest concentration of reactors in the world.[48]

Experimental Breeder Reactor Number 1 in Idaho, the first reactor to provide electricity for public use.

What is now Idaho National Laboratory in southeastern Idaho began its life as a U.S. government artillery test range in the 1940s. Shortly after the Japanese attacked Pearl Harbor, the U.S. military needed a safe location for performing maintenance on the Navys most powerful turreted guns (16-inch diameter). The guns were brought in via rail to near Pocatello, Idaho, to be re-sleeved, rifled and tested.[49]

In 1949, the U.S. Atomic Energy Commission established the National Reactor Testing Station or NRTS at the site.

As the Navy began to focus on post-World War II threats, the types of projects worked on in the Idaho desert changed, too. Perhaps the most well-known was the building of the prototype reactor for the worlds first nuclear-powered submarine, the USS Nautilus

In the early afternoon of Dec. 20, 1951, Argonne National Laboratory scientist Walter Zinn and a small crew of assistants witnessed a row of four light bulbs light up in a nondescript brick building in the eastern Idaho desert.[50] Electricity from a generator connected to Experimental Breeder Reactor-I (EBR-I) flowed through them. This was the first time that a usable amount of electrical power had ever been generated from nuclear fission.

Only days afterward, the reactor produced all the electricity needed for the entire EBR complex.[51] One ton of natural uranium can produce more than 40 million kilowatt-hours of electricity this is equivalent to burning 16,000 tons of coal or 80,000 barrels of oil.[52]

More central to EBR-Is purpose than just generating electricity, however, was its role in proving that a reactor could create more nuclear fuel as a byproduct than it consumed during operation. In 1953, tests verified that this was the case.[50] The site of this event is memorialized as a Registered National Historic Landmark, open to the public every day Memorial Day through Labor Day.

On January 3, 1961, the only fatal nuclear reactor incident in the U.S. occurred at the NRTS. An experimental reactor called SL-1 (Stationary Low-Power Plant Number 1) was destroyed when a control rod was pulled too far out of the reactor, leading to core meltdown and a steam explosion. The reactor vessel jumped up 9feet 1inch (2.77m).[53] The concussion and blast killed all three military enlisted personnel working on the reactor. Due to the extensive radioactive isotope contamination, all three were buried in lead coffins. The events are the subject of two books, one published in 2003, Idaho Falls: The untold story of America's first nuclear accident,[54] and another, Atomic America: How a Deadly Explosion and a Feared Admiral Changed the Course of Nuclear History, published in 2009.[53]

On November 8, 2011 in the Zero Power Physics Reactor (ZPPR) in the afternoon a container leaked "plutonium-related" materials, when it was opened by one of the workers. All 17 workers at the incident were immediately taken to have testing done by the Idaho Cleanup Project in the form of Whole Body Counts (scans the body for any internal radiation exposure) and were required to submit urine and fecal samples to further test for internal radioisotopes . Six of them proved to be exposed to "low-level-radiation", two of them fairly extensively. All workers were kept under close observation afterwards with repeated Whole Body Counts and urine and fecal sampling. The Idaho National Laboratory insisted that no radioactivity leaked outside the facility.[55]

From 1969 to 1994, Argonne National Laboratorys EBR-II produced nearly half of the electricity needed for test site operations.

In 1964, Experimental Breeder Reactor II and the nearby Fuel Conditioning Facility proved the concept of fuel recycling and passive safety characteristics. So-called passive safety includes systems that rely on natural physics laws such as gravity rather than systems that require mechanical or human intervention.

In a landmark test on April 3, 1986, such systems in EBR-II demonstrated that nuclear power plants could be designed to be inherently safe from severe accidents.

De-commissioning of EBR-II began in October 1994 with the removal of the 637 fuel assemblies.[56]

The worlds first Loss-of-Fluid-Test reactor started up at INL on March 12, 1976. It repeatedly simulated loss-of-coolant accidents that could potentially occur in commercial nuclear power plants. Many safety designs for reactors around the world are based on these tests. LOFT experiments helped accident recovery efforts after the Three Mile Island accident in 1979.[57]

In 1949, an area of the fringe of the NRTS property named "Test Area North", or TAN, was developed by the U.S. Air Force and the Atomic Energy Commission to support the Aircraft Nuclear Propulsion program's attempt to develop a nuclear-powered aircraft. The programs' Heat Transfer Reactor Experiments (HTRE) were conducted here in 1955 by contractor General Electric, and were a series of tests to develop a system of transferring reactor-heated air to a modified General Electric J47 jet engine. The planned aircraft, the Convair X-6, was to be test flown at TAN, and a large hangar with radiation shielding was built on the site. The program was cancelled, however, before the accompanying 15,000-foot (4,600m) runway was built.

In the early 1950s, the very first full-scale prototype nuclear plant for shipboard use, called S1W Prototype, was constructed to test the feasibility of using nuclear power aboard submarines. It was the predecessor to a similar nuclear plant of S2W design installed in the first nuclear-powered ship, the submarine USSNautilus(SSN-571). Later, two more prototype plant facilities, A1W and S5G, were built at this location called the Naval Reactors Facility (NRF for short). There is also an Expended Core Facility (ECF for short) also at NRF as well as administrative buildings/facilities. NRF's chemistry lab was located at the S1W prototype. By now, the prototype plants for shipboard use development have been shut down. Only the Expended Core Facility / Dry Storage Area is in use.

When the nuclear industry was just getting started in the early 1950s, it was difficult to predict exactly how different kinds of metals and other materials would be affected by being used in a reactor for prolonged periods of time. MTR was a research reactor jointly designed by Argonne and Oak Ridge National Laboratories that operated until 1970 and provided important data, helping researchers make nuclear power reactors safer and longer lasting.[58]

The Boiling Water Reactors (BORAX) experiments were five reactors built between 1953 and 1964 by Argonne National Laboratory. They proved that the boiling water concept was a feasible design for an electricity-producing nuclear reactor. One of the BORAX reactors (III) was also the first in the world to power a city (Arco, Idaho) on July 17, 1955.[59][60]

The Idaho Chemical Processing Plant chemically processed material from used reactor cores to recover reusable nuclear material. It is now called the Idaho Nuclear Technology and Engineering Center.

The Materials Test Area tested materials' exposure to reactor conditions. The Materials Test Area is part of the Advanced Test Reactor Complex.

Central Facilities Area where whole body counts for radioactivity are done for INL employees.

Idaho National Laboratorys Advanced Vehicle Testing Activity (AVTA) about plug-in hybrids (PHEVs).

The New York Times reported in 2005 that a reactor at INL would be used to manufacture plutonium-238, most of it for classified national security purposes.[61] This isotope is known for its intense alpha decay, which is useful in making extremely long-lived power sources such as radioisotope thermoelectric generators (RTG)s for deep space probes and heart pacemaker batteries. INL has 52 reactors, three of which are reportedly still operating (see list of nuclear reactors). The Idaho State Journal reported that the batteries would be used for a voyage to Jupiter's moons and the New Horizons trip to Pluto.[62]

Coordinates: 433200N 1125641W / 43.53333N 112.94472W / 43.53333; -112.94472

View original post here:
Idaho National Laboratory - Wikipedia

Posted in Idaho Stem Cells | Comments Off on Idaho National Laboratory – Wikipedia

Sickle Cell Trait – sickle. bwh .harvard.edu

Posted: November 30, 2016 at 6:41 pm

John Kark, M.D. (formerly of the Uniformed Services University of the Health Sciences, Bethesda, MD.) Howard Universty School of Medicine Center for Sickle Cell Disease 2121 Georgia Avenue Washington, D.C. 20059 revised December 20, 2000 Introduction Sickle cell trait usually is not regarded as a disease state because it has complications that are either uncommon or mild. Nevertheless, under unusual circumstances serious morbidity or mortality can result from complications related to polymerization of deoxy-hemoglobin S. Such problems include increased urinary tract infection in women, gross hematuria, complications of hyphema, splenic infarction with altitude hypoxia or exercise, and life-threatening complications of exercise, exertional heat illness (exertional rhabdomyolysis, heat stroke, or renal failure) or idiopathic sudden death (1-4). Pathologic processes that cause hypoxia, acidosis, dehydration, hyperosmolality, hypothermia, or elevated erythrocyte 2,3-DPG can transform silent sickle cell trait into a syndrome resembling sickle cell disease with vaso-occlusion due to rigid erythrocytes. Compound heterozygous sickle cell disease can be mistaken as uncomplicated sickle cell trait, particularly when an unusual globin variant is involved.

In addition some disease associations have been noted with sickle cell trait which might not result from polymerization of hemoglobin S but from linkage to a different gene mutation. The association of hemoglobin S with cases of renal medullary carcinoma, early end stage renal failure in autosomal dominant polycystic kidney disease, and surrogate end points for pulmonary embolism are not necessarily the result of hemoglobin S polymerization. Complications from sickle cell trait are important because about three million people in the United States have this genotype, about 40 to 50 times the number with sickle cell disease.

People with uncomplicated sickle cell trait have a normal blood examination as assessed by conventional clinical methods, including normal red cell morphology, indices, reticulocyte counts, and red blood cell survival by chromium labeling. Conventional methods of detecting hemolysis are negative, such as measurements of serum haptoglobin, bilirubin, and LDH. Erythrocyte density distribution is normal, adherence to endothelium is not increased, altered membrane lipids and proteins are not detectable, cytoplasmic inside-out vesicles with high calcium content are absent, and permanently distorted erythrocytes are not observed.

When blood is drawn with anaerobic technique into a syringe with dilute buffered glutaraldehyde one obtains an accurate picture of circulating erythrocytes in vivo (the Sherman test). No sickled cells are observed at rest, but exercise to exhaustion at sea level regularly induces mild levels of reversible sickling in peripheral venous blood (less than 1%). Exposure to altitude hypoxia will progressively increase the extent of sickling observed with sickle cell trait from 2% at 4,050 ft. to 8.5% at 13,123 ft. Hypobaric chamber exposures used for military aviation training, involving hypoxic exposures simulating 10,000 to 25,000 ft from ninety to six minutes, did not cause hemolysis in subjects with uncomplicated sickle cell trait (3).

Determination that a clinical syndrome is due to sickle cell trait rather than a subtle form of sickle cell disease is difficult. Reversible sickling and unsickling of erythrocytes (reflecting the rapid formation and dissolution of deoxy-hemoglobin S polymers) takes place in seconds. Hence, the presence or absence of intravascular sickled erythrocytes in tissue specimens depends upon the degree of oxygenation of the sample just before fixation and only has clinical relevance if fixation occurred at oxygen tensions identical to those extant during generation of primary lesions. Agonal hypoxemia causes artifactual intravascular sickling. Conversely, blood samples smeared in room air and then fixed will show artifactual unsickling. One cannot determine the role of hemoglobin S in clinical events from the presence or absence of intravascular sickling in blood samples, biopsy specimens, or autopsy specimens unless these were rapidly fixed at physiologic oxygen tension.

While fatal intravascular sickling with extensive microvascular obstruction could theoretical result from sickle cell trait, such an event cannot be demonstrated by histologic examination at autopsy. If a clinical event is not specific for hemoglobin S, one may need to show that the complication occurs significantly more often in people with sickle cell trait relative to a control group. Such an association does not prove cause. Stronger evidence that polymerization of hemoglobin S causes a problem is demonstration of relative protection by alpha thalassemia.

The common African polymorphism causing alpha thalassemia is the product of a prior mismatched cross over event which creates chromosome 16 expressing only one of the two alpha globins and a chromosome 16 carrying three alpha globin exons. Loss of one or two alpha globin genes decreases the fraction of hemoglobin S and produces obvious microcytosis. Anemia is absent or mild.

Examination of maximal urinary concentrating ability in people with sickle cell trait relative to alpha globin gene number demonstrated that one or two alpha globin gene deletions were associated with better preserved renal function (5). In other words the less hemoglobin S that was present, the less renal function that was lost. This implied a significant role of polymerized hemoglobin S in the pathogenesis of renal isosthenuria (see below). In some instances the anatomic lesions due to sickle cell trait are so distinct that a relationship to polymerization of Hb S can be reasonably inferred. Such complications of sickle cell trait include glaucoma or recurrence after treatment for hyphema and splenic infarction in the absence of primary trauma, infection, inflammation or tumor in the spleen.

People with sickle cell trait often experience subclinical tissue infarction from microvascular obstruction by rigid erythrocytes. Most people with sickle cell trait develop microscopic infarction of the renal medulla because the extreme hypoxemia, hypertonicity, acidosis, and hyperthermia of arterial blood passing through the long vasa recta of the renal medulla promote polymerization of deoxy-hemoglobin S (6). Flow through these vessels requires more than ten seconds, providing an unusually long exposure time for polymerization of hemoglobin S. Cumulative focal lesions result in loss of maximal urine concentrating ability which is progressive with age and develops in most adults with sickle cell trait (3, 6). The functional defect limits urine concentration to approximately the osmolality of serum, causing isosthenuria rather than hyposthenuria. In people with sickle cell trait urine osmolality can usually reach values higher than plasma during overnight dehydration (400 to 800 mOsmol). Although one may speculate that this lesion might predispose to development of mild exertional heat illness (EHI) during exercise in hot weather, clinically significant problems related to this deficit have not been demonstrated. Necrosis of the renal papillae can result in hematuria, which is usually microscopic. Gross hematuria is occasionally provoked by heavy exercise or occurs spontaneously.

An important potential complication of sickle cell trait is unexpected exercise-related death (ERD). The validity of this association aroused heated controversy (4). The possibility that previously healthy young people with sickle cell trait might suffer increased mortality from exercise was first suggested by observations of enlisted recruits in US Armed Forces basic training. A military trainee with Hb AS suffered exercise related hypernatremia during physical training in the field. He only survived a critical illness that included acute renal failure because of dialysis (8). During a single summer, there were four exercise-related deaths among recruits at Fort Bliss, all of whom were black and had sickle cell trait, while no recruits with normal hemoglobin died. Only 1.5% of these recruits had sickle cell trait. The authors suggested a significant risk association with sickle cell trait (8).

Twelve cases of natural exercise-related death (ERD) among apparently healthy young men with Hb AS were reported by 1981. These deaths were predominantly due to exertional rhabdomyolysis, although some were sudden idiopathic deaths with cardiopulmonary arrest, associated in two cases with hyperkalemia. Identical presentations were observed in recruits without Hb S. There is no direct proof that sickle cell trait contributed to ERD through microvascular obstruction by rigid erythrocytes. There is little evidence that these deaths involve the typical acute complications of sickle cell disease, such as acute focal infarction of the spleen, kidneys, lungs, bone, retina, or brain, sudden extensive sequestration of blood in the spleen or liver, or overwhelming infection with encapsulated bacteria.

In 1981 we embarked on studies of exercise-related death among US military enlisted recruits in basic training which took advantage of the potential for accurate epidemiologic analysis. Large exercising populations of apparently healthy young adult recruits were enumerated with an accuracy greater than 96% in a database describing each individual. Because of medical, legal, and military command concerns, each recruit death has been investigated in detail, with a full autopsy and toxicology, clinical records, and eyewitness accounts. We added assessment of these materials by experts in forensic pathology, cardiovascular pathology, and internal medicine. Selection bias was eliminated by obtaining all cases of exercise-related death in the study population. In contrast, the frequently cited surveys of civilian athlete deaths usually required selection of those cases identified as sudden deaths, often were selected from poorly defined or poorly measured athlete populations, and athlete cases frequently lack a complete autopsy, toxicology, or full eyewitness accounts (14).

We performed complete cohort study of ERD among the 2.1 million people who entered US Armed Forces basic enlisted military training during the five years, 1977-1981 (9). The population was dived into black and non-black groups to estimate the fraction with Hb AS from published surveys. Prevalence of Hb AS was 8% among 20,600 black recruits (10) and 0.046% among 57,600 non-black recruits (11). There were 37,300 black recruits with Hb AS, 1,300 non-black recruits with Hb AS, 429,000 black recruits without Hb AS, and 1,620,000 non-black recruits without Hb S. Forty-one exercise-related deaths occurred. Hb AS was only found among natural deaths. Risk ratios were examined among the black recruits, ignoring the small number of non-blacks with Hb AS.

The relative risk of ERD explained by preexisting disease (largely silent heart disease) was 2.3 for Hb AS , but this was not statistically significant. The relative risk of ERD unexplained by preexisting disease was 28 for Hb AS. This was highly significant with p less than one per thousand. The relative risk ratio has since been corrected to 30 (3). If one eliminates restrictions by race and cause of ERD, the risk of exercise-related death for sickle cell trait was 28-fold. The excess ERDs with sickle cell trait seemed to result from the immediate stress of exercise. About 50% of cases resulted from exertional heat illness and the remaining cases were idiopathic sudden deaths (ISD). Clinical features and distribution of cases between EHI and ISD did not differ by the presence or absence of hemoglobin S, except that rhabdomyolysis was the predominant form of EHI among cases with sickle cell trait (3).

We examined the effect of age on risk of ERD unexplained by preexisting disease. There was an eight-fold increase in mortality going from age 17-18 to age 28-29 among recruits with Hb AS but no such trend for recruits without sickle cell trait (3, 9). This difference in effect of age suggests that there may be a difference in pathogenesis of death depending on the presence or absence of hemoglobin S. This effect might be due to renal papillary necrosis from Hb AS, a lesion increasing linearly in severity with age and present in at least 80% of recruits (figure 1 in 3, 6). The resulting deficit in renal concentrating ability might predispose that person toward more severe EHI since obligatory loss of free water might increase the hyperosmolar state important in the pathogenesis of EHI.

We were surprised by the high excess mortality associated with sickle cell trait. It is often said that the absolute risk of mortality with sickle cell trait we reported was low (12, 13). This excess mortality was one per three thousand recruits with sickle cell trait or one death per 60 to 90,000 person-hours of exercise equivalent to middle distance running. This mortality rate for 18 year old recruits is about 4 to 7 times higher than the mortality observed from artherosclerosis among middle aged runners: one death per 400,000 hours of running (14). Other population surveys of sickle cell trait have shown only mild effects of trait on hospitalization rates and none on mortality rates (3). Whereas our survey observed 5,000 person-years of exposure (38,600 people with Hb AS for a median of 8 weeks exposure) (9), other surveys of young adults with sickle cell trait examined exposures two to four logs smaller.

Heller et al. examined hospital admissions over three years for 4,900 veterans with Hb AS with a median age of 49 years and no time for follow-up (15). This older population would not be expected to engage in conditioning exercise and therefore would not be subject to a comparable risk of exercise related-death. An important study of Navy enlisted members with sickle cell trait examined 599 recruits with sickle cell trait during a four year tour of duty, about 2,400 person-years of exposure (16). Exercise-related mortality rates with Hb AS are at least ten-fold lower for military members than recruits, making this study too small to identify mortality related to sickle cell trait (4).

We subsequently determined ERD rates in US Armed Forces basic training for 98,800 black recruits with Hb AS and 1.14 million black recruits without Hb S. We found that Hb AS was associated with a 21-fold higher relative risk of ERD unexplained by preexisting disease (17, table 6). This implies an excess mortality with sickle cell trait of one per 5,500 recruits exposed to eight weeks of training. The reduction in risk is explained by intervention to reduce mortality for a subset of these (see below).

All but one of the large autopsy series of exercise-related deaths among athletes have shown that exertional heat illness accounted for less than 1% of cases and that idiopathic sudden death accounted for between 5 and 12% of such deaths (3, 17). However, our survey of 41 recruit ERDs, demonstrated that non-sudden exertional heat illness deaths and idiopathic sudden death each accounted for about one-third of ERD (3, 17). Only five percent of sudden deaths, whether explained by preexisting cardiac disease or idiopathic, were appropriately screened by determination of body temperature near death and by serum and urine chemistry studies to exclude exertional heat illness. It seemed possible that EHI contributed to a much larger fraction of recruit deaths than was found in most of the autopsy studies of ERD of civilian athletes.

We have reviewed 55 cases of unexpected ERD with sickle cell trait (3). At least two-thirds occur under conditions of high risk for EHI. Most deaths were non-sudden. Those few cases of sudden ERD which were properly examined demonstrate hyperthermia or chemical abnormalities diagnostic of acute EHI. In our recruit cohort study of 94 consecutive recruit ERDs at least two-thirds of ISD (with or without hemoglobin S) resulted from middle distance running during the hot season but early in the morning when the immediate heat stress was considered safe (3). Unrecognized exertional EHI might have contributed to these deaths.

Current military standards were designed in the 1950's for recruits whose physical conditioning was predominantly marching at 6 METS (METS are units of oxygen consumption for a given weight over a minute. Moderate walking is 3 METS, cycling is 6 METS). Since the 1970's recruit conditioning has been predominantly middle distance running at 12-14 METS, implying a need for altered activity at lower heat index levels. Substantial risk of EHI might also result from prior-day heat exposure, which is not considered a risk factor for heat illness by current military or sports medicine standards. We examined these issues in a ten year cohort study of Marine recruits (18). We related rates of EHI from 1,454 consecutive cases of non-fatal exertional heat illness to hourly values for the wet bulb black globe temperature index (WBGT), the heat index best related to physiologic response to exercise in heat.

This study demonstrated that prior-day exposures and heat stress at WBGT values between 70 and 84F were important determinants of rates of EHI among recruits (18). A preliminary analysis of 94 consecutive recruit ERD cases was performed, using WBGT values for the 24 hours prior to presentation of EHI to identify conditions in which the risk of EHI was increased at least 15-fold. This study suggests an association between high-risk of exertional heat illness from environmental exposure and ERD with sickle cell trait (all ISD cases), a substantial association for ISD without hemoglobin S (54% of cases), and sudden explained cardiac death without hemoglobin S (42% of cases versus 11% of recruit deaths unrelated to EHI) (19). We have been able to describe in detail eight cases in which sudden death occurred with acute exertional heat stroke or rhabdomyolysis. In a small cohort study at one training center, we demonstrated a strong relation between severe exertional heat illness and life-threatening or fatal cardiovascular complications for recruits without hemoglobin S (3).

The ultimate test of the hypothesis that unrecognized EHI contributes to ISD (especially among people with sickle cell trait) would be to conduct effective prevention of EHI during exercise and observe an appropriate reduction in mortality. In February 1982 we proposed stricter rules for drill instructors in order to correct the major deficiencies in preventative measures for EHI which we noted at most recruit training centers during 1981. These rules provided prevention based on 30 to 60 minute measures of the WBGT at the actual exercise site and direct observation that each recruit was drinking the amount of water recommended to prevent EHI. This intervention was applied to all trainees and did not require prior identification or different management of individuals with sickle cell trait. We tested the effect of this intervention on ERD rates prospectively during the next ten years of training (1982-1991). Participating centers adhered to this intervention while training 2.3 million recruits and non-participating centers did not adopt these unproven recommendations while training 1.2 million recruits. Preliminary analysis of this trial has been presented (20).

Based on the ERD rates observed in 1977-1981 (3, 9), we predicted 15 deaths with sickle cell trait at participating centers. No deaths were observed in the training of 40,000 recruits with sickle cell trait. There was a trend toward better survival among recruits without hemoglobin S (19 deaths predicted but only 11 observed). Among non-participating centers there was no significant difference between predicted and observed deaths (14 each) regardless of hemoglobin type. These data support the view that preventable unrecognized exertional heat illness is the predominant factor causing exercise-related deaths with sickle cell trait and may be a substantial factor contributing to such deaths in recruits without hemoglobin S. This approach appeared able to prevent excess mortality with sickle cell trait in recruit basic training. This study has not undergone peer review nor have the full details of the trial been published.

The question remains of whether or not excess ERD rates with sickle cell trait are caused by polymerization of hemoglobin S. We have attempted to determine whether alpha-thalassemia is protective for ERD with sickle cell trait. We sought well-defined cases of fatal or life-threatening complications of exercise in healthy young adults with sickle trait and a report of quantitative hemoglobin electrophoresis. Preliminary analysis of 33 cases showed that the frequency of alpha-thalassemia in these cases was more than ten-fold below the expected value for unselected African-Americans (3, 21). Current analysis of 44 cases substantiates this. We conclude that a low fraction of hemoglobin S must be protective for ERD with sickle cell trait. Polymerization of hemoglobin S must be a necessary part of pathogenesis of excess fatalities with sickle cell trait. The possibility remains that additional mutations genetically linked to the beta globin gene are critical and define a susceptible subset of people with sickle cell trait.

Important risk factors for EHI which have been associated with ERD of young adults with sickle cell trait include inadequate hydration, environmental heat stress with a WBGT of at least 75F during the preceding 24 hours (18), heat retaining clothing, sustained heroic effort above customary activity, incomplete acclimation to heat, obesity with poor exercise fitness (22), inadequate sleep, and delay in recognition and treatment of EHI. The majority of cases were among recruits in basic training, and very few cases among permanent military members. About one-third of published cases resulted from civilian athletic or physical training programs. We are unaware of cases resulting from heavy work. The largest group of American athletes reported with sickle cell trait and fatal EHI was football players during preseason training. It is plausible that this situation combines risk factors from high environmental heat stress, poor acclimation, poor conditioning, heat retaining clothing, and a higher frequency of sustained metabolic exercise.

An important question is why ERD rates are more than ten-fold lower among military members than in entry training. While it is possible that highly susceptible individuals are removed by discharge or death in entry training, circumstances may be more dangerous during entry training. In support of this view, many of the fatalities during military service have come from demanding conditioning programs in specialized training schools for military members. The increased risk with age noted for recruits does not seem to apply to military members, with only two military and one published civilian case aged more than 30 years. We believe that risk of unexpected ERD is largely confined to periods of intense conditioning for a new form of exercise or a sustained event at a level of performance for which the individual is unprepared. There are many reports indicating no increased morbidity or mortality for competitive professional athletes with sickle cell trait (3). Professional athletes remain fit during the off-season and seldom have to go through conditioning at an intensity comparable to military basic training. Further, water for hydration is readily available during athletic events.

Our recommendations for safe exercise by individuals with sickle cell trait are based upon the premise that the predominant cause of excess morbidity and mortality is preventable exertional heat illness. At least half of these cases were proven to suffer from acute exertional heat illness, with rhabdomyolysis the predominant component. The other half of cases died suddenly without a clear etiology, but with evidence for increased risk of unrecognized EHI when such evidence was sought. The controlled study supporting this view has not undergone peer review and publication.

Effective prevention of EHI during demanding physical conditioning requires following measures similar to those used by recruits and distance runners (3, 18-20). Performance levels should be built up gradually, avoiding severe muscle pain. Training should cease and restart gradually when substantial myalgia occurs. Adequate hydration with increased water intake rising with environmental heat stress is essential. In the evening of any hot day with a WBGT value above 75F, the athlete should be sure to ingest adequate amounts of salt and potassium to replace sweat losses and water to replace fluid deficits. We recommend checking the color of the first AM urine in a clear plastic cup as an easy method to identify people who are dehydrated from prior day heat exposure if measurement of urine specific gravity is not readily available. Those with darker urine can drink an additional pint or quart of water before starting exercise. Athletes in a demanding training program can keep a log of daily weights from the same scale on waking and on going to sleep.

Over-hydration is possible with consequent hyponatremia, seizures, and death. Oral hydration should not exceed one quart per hour or 12 quarts per day without monitoring of blood chemistries. Patients with muscle cramps require additional salt, which can be taken orally as two teaspoons of salt in a quart of water or intravenously as half-normal or normal saline. During sustained exercise, such as marching, middle to long distance running, basketball, and soccer, athletes should drink water at intervals of approximately 15-20 minutes.

Sodium and potassium replacement with meals avoids aggravating the common trend toward hypernatremia during exercise in heat. One should be careful to avoid sustained full intensity efforts lasting more than two minutes that require at least six METS, with special attention to exercise at full competitive intensity or that requires over ten METS for more than two minutes. Levels of activity should be adjusted for the WBGT level at the exercise site measured every 30-60 minutes. At the same time the fraction of time spent at rest and the minimal level of hourly hydration should increase with rising WBGT.

The level of heat stress is affected by the extent to which clothing permits heat loss and blocks radiant sunlight. High metabolic activity should be conducted in loose, light clothing during hot weather, with appropriate protection from radiant sunlight, such as head cover, during lesser activity. Rapid treatment in the field and during transport to a hospital is the best was to minimize the severity of exertional heat illness. Demanding physical conditioning is safer when conducted with an experienced trainer or medical personnel.

The athlete with sickle cell trait should understand the non-specific early warning symptoms and signs of EHI and obtain medical advice immediately. Common sense measures to optimize hydration and cooling should be started as soon as EHI is suspected. People with sickle cell trait should also be aware of the presenting symptoms and signs of hematuria and splenic infarction, both of which can occasionally occur as a consequence of heavy exercise. Individuals with sickle cell trait are potentially at greater risk with higher metabolic activity, longer periods of sustained effort, and exposure to high altitude.

Splenic infarction from sickle cell trait is more common with exercise at high altitude but has occurred with altitude exposure at rest or with exercise at sea level (3). Since there is no means to acclimate to this risk we advise against high altitude exposure and sustained exercise at an altitude greater than 7,000 feet, but there is contrary evidence in the literature. Many individuals with sickle cell trait have participated at professional and international levels of sport, including reports from the Olympic competition in Mexico City and high altitude long distance running in the Cameroon. Theoretically consistent maintenance of conditioning and consistent adjustments to minimize EHI permit continued safe levels of participation.

A relatively common clinical problem is what advice to give a person with sickle cell trait who has experienced exertional heat illness. I recommend permanent avoidance of physical activity at a comparable level after a single occurrence of severe EHI (heat stroke, or rhabdomyolysis with renal failure, or isolated renal failure). The level of risk following serious EHI has never been adequately measured, but expert opinions for patients with normal hemoglobin is that an increased risk of EHI is very likely for at least three to six months and may exist for years.

A more difficult problem is how to advise someone with Hb AS who develops asymptomatic elevations of serum muscle enzymes or myoglobinuria with a particular activity. Often this problem will resolve if the activity is conducted with attention to maintaining a low risk of EHI. However, several case reports described fatal rhabdomyolysis from middle distance running following such warning events. After a single warning event, one can cautiously condition the patient to a lower maximal activity, ensure that circumstances are optimal to avoid EHI and retest serum muscle enzyme levels 24 hours post-exercise. If inappropriate elevation of muscle enzymes persists, we advise that the person permanently limit their maximal physical activity to a level which does not raise the serum muscle enzymes. The physician should consider the possibility of an inherited metabolic disorder contributing to unexpected elevation of serum muscle enzymes.

The spleen is unusually susceptible to vaso-occlusion related to hemoglobin S polymerization and red cell deformation. When persons with hemoglobin S are exposed acutely to high altitude hypoxia, the spleen is the organ most consistently injured by micro-vascular obstruction. Splenic infarction usually presents as severe abdominal pain localizing within a few hours to the left upper quadrant, accompanied by nausea and vomiting. Splinting of the left hemithorax, left pleural effusion, and atelectasis of the left lung often follow. A tender enlarged spleen often becomes palpable. Fever, leukocytosis, and an acute elevation of serum LDH level occur during the first 72 hours, out of proportion to serum CK, AST, or ALT levels. Splenic infarcts are best imaged by CT scan, which usually shows a few large regions of hemorrhage of variable size. Often small hemorrhages collect outside the splenic capsule.

While this appearance can be mimicked by a number of other processes, such a pattern of necrosis with acute onset is unlikely when the precipitating disorder is not obvious (for example, myeloid metaplasia). One exception is traumatic hemorrhage, which can progress over months or years, causing slow enlargement of the spleen and even erosion of adjacent ribs. Splenic infarction is readily differentiated from the early lesions of DIC, which are tiny foci of hemorrhagic necrosis diffusely scattered throughout the spleen. Prolonged DIC may produce large areas of hemorrhage, mimicking infarction from sickle erythrocytes. Liver-spleen scan with sulfur colloid usually demonstrates decreased perfusion of large regions of the spleen but are not as sensitive as the CT scan. Splenic infarction with sickle cell trait is usually self-limited, resolving in 10 to 21 days, and rarely requiring surgical intervention.

Non-traumatic splenic infarction following altitude hypoxia is most likely to occur in people with sickle cell disease and an enlarged and functional spleen prior to exposure. Such patients usually have hemoglobin SC or hemoglobin S/+ -thalassemia genotypes rather than hemoglobin SS (3). Those with hemoglobin SS genotype have an atrophic, fibrotic spleen and are relatively protected from splenic infarction. These patients do, however, develop pain crisis in other locations. Splenic infarction with sickle cell disease occurs after shorter and milder exposures and is often more severe than with sickle cell trait. Presentation with severe anemia or progression of splenectomy are much more likely with sickle cell disease. People with sickle cell trait account for more cases of splenic infarction by dint of their larger number. The per capita incidence of splenic infarction is lower than with sickle cell disease. Sickle cell trait does not produce gradual chronic fibrosis or gradual splenomegaly. Rupture of the spleen requiring emergency splenectomy has been described twice in people with sickle cell trait (26, 27). Two patients with sickle cell trait and spherocytosis required splenectomy because of severe sequestration crisis (28).

When reviewing cases of splenic infarction attributed to sickle cell trait, it is important to confirm that the patient did not have a form of sickle cell disease. Strict criteria to identify sickle cell trait as a cause of this syndrome are: a positive phosphate precipitation test or slide test for sickling (establishing the presence of hemoglobin S), a hemoglobin electrophoresis pattern consistent with sickle cell trait (e.g. more hemoglobin A than hemoglobin S and normal levels of trace hemoglobins), and a normal erythrocyte morphology, hemoglobin concentration, hematocrit, erythrocyte indices, and reticulocyte count when not acutely ill. Published case reports frequently lack comprehensive data, especially descriptions of erythrocyte morphology, splenic pathology and demonstration of recovery from acute anemia and an explanation of abnormal red cell indices, morphology, or acute elevations of reticulocyte counts. In adults with sickle cell disease there may be congestion and hemorrhage around terminal arterioles, fibrosis and thickening of terminal arteriolar walls, discreet infarcts, and scattered siderofibrotic and calcified nodules, the characteristic chronic lesion left at the end of hemorrhage or infarction. While these changes will follow symptomatic infarction with sickle cell trait, if there is no gross scarring and fibrosis and no siderofibrotic bodies in the spleen of an adult known to have hemoglobin S, one can reasonably conclude that the patient did not have sickle cell disease.

We summarized the published case reports of splenic infarction with sickle cell trait available to April 1994 (3, Table 8). Since then six published cases fulfill criteria for adequately documented cases of splenic infarction due to sickle cell trait and one did not. Two cases were added from my practice, for a total of forty-seven documented cases. Eleven of these cases exhibited acute recticulocytosis attributed to splenic hemorrhage and sequestration. Seven patients had abnormal erythrocyte morphology implying an additional red cell abnormality. Two cases exhibited an elevated hemoglobin concentration. Recovery from anemia was unconfirmed in the absence of follow-up studies for 13 patients.

Fifteen cases resulted from exposure to hypoxia at rest in aircraft and one additional case involved exposure in aircraft followed by exercise on the ground. Only one case involved a crew member, who might have been physically active during flight. No published cases have involved pilots. Twelve of the 25 cases with splenic infarction on the ground were related to exercise. Four case reports did not clearly exclude exercise at altitude. Two patients had hypoxemia from disease rather than ambient oxygen tension. Splenic infarction was reported in at least ten patients with sickle cell trait who were exposed to no substantial altitude hypoxia, denied any exercise near the onset of symptoms, and were not known to have any defect in blood oxygenation. Two patients had acute hypoxemia in the hospital attributed to the effects of respiratory splinting, but without values after recovery to exclude chronic hypoxemia. Splenic infarction occurred previously with hypoxic exposure in ten cases.

The first adequately documented association of sickle cell trait with splenic infarction involved the large population of black servicemen flying in unpressurized aircraft during the Korean War. Subsequently many cases of splenic infarction were reported from Lake Tahoe and other resorts in the Rocky Mountains. These cases were often of Mediterranean or mixed ancestry. The possibility that white individuals with sickle cell trait are at greater risk of splenic infarction was suggested because of their predominance among cases occurring on the ground (3, 29). Among 32 patients with sickle cell trait whose splenic infarction occurred while on the ground, at least 24 had white ancestry (75%). Since roughly four percent of Americans with sickle cell trait are non-black, one would expect only one non-black patient. This difference was significant with p less than 0.01. Among 15 patients with splenic infarcts in aircraft, two were white. Perhaps even more striking, only four out of the 47 reported cases of splenic infarction with sickle cell trait were female. It is unclear whether the high association of risk with male gender or with non-black ancestry is due to undefined additional factors or is partially explained by the unusual populations from which cases are reported. High normal levels of MCV may be a risk factor for splenic infarction, possibly as a marker for red cell membrane defects which are thought by some to increase risk of splenic sequestration and splenic hemorrhage (3). It is surprising that splenic infarction has not been detected among cases of non-sudden death from exertional heat illness during heavy exercise, as reported for twenty of our patients (3). These observations support the view of Lemeul W. Diggs that sickling might not initiate the serious complications of exercise but may increase the severity when serious episodes occur (30).

The effect of a-thalassemia on frequency of complications related to sickle cell trait was discussed above. We performed an estimate of the frequency of alpha-thalassemia among cases of splenic infarction with sickle cell trait, using a hemoglobin S fraction less than 35% as a marker among the 33 patients with quantitative hemoglobin electrophoresis known to us. Three patients had hemoglobin fractions less than 35% versus the ten expected. This demonstrates approximately a three-fold protective effect of alpha-thalassemia trait, and implies that pathogenesis of splenic infarction involves polymerization of hemoglobin S.

The frequency of hematuria with sickle cell trait from renal papillary necrosis has been accurately measured in a single large study of elderly patients in Veterans Hospitals (15). Patients with sickle cell trait had a 4% admission rate for hematuria , a significantly higher rate than the 2% admission rate for patients with normal hemoglobin. The absolute rate of substantial hematuria requiring admission was probably higher than would be the case for a population not selected by hospitalization. It is reasonable to conclude that sickle cell trait results in an approximate doubling of the incidence of hematuria from an unknown absolute incidence of 2% or less of hospitalizations. This implies that hematuria in people with sickle cell trait is often unrelated to sickling or papillary necrosis.

Patients with hematuria should be evaluated by an urologist, who will perform imaging studies and obtain tissue as needed to exclude structural lesions such as neoplasms and stones and correct any related problems with flow of urine from the calyces to the urethra. Hematuria from a coagulopathy should be considered. A few patients have been described whose hematuria was associated with otherwise asymptomatic von Willebrand's disease. A small number of case reports suggest that patients with sickle cell trait and hematuria who have von Willebrand's disease may have responded to infusion of DDAVP or cryoprecipitate. Efficacy has not been demonstrated by controlled studies. Since such therapy is low risk and has been effective for a few patients, one could consider screening for von Willebrand Factor deficiency. In the absence of controlled data for prevalence and effectiveness of therapy, most physicians only search for von Willenbrand factor deficiency if the prior history or the family history suggests a bleeding disorder.

Gross hematuria producing anemia and requiring blood transfusions has an unpredictable course. The problem can persist for weeks to months with many recurrent episodes. Occasionally bleeding becomes so profuse that anemia is life-threatening. Episodes of severe bleeding are more common in men than in women. Most are unilateral with the left kidney involved more frequently than the right, while 10% are bilateral. Spontaneous resolution without any treatment occurs frequently, making the evaluation of any form of treatment difficult in the absence of controls. Patients with serious hematuria usually have involvement of multiple papillae in both kidneys, with a high risk of eventual bleeding on the opposite side. Nephrectomy should be avoided if at all possible.

Evaluation of hematuria routinely includes an IVP, ultrasound, and CT scan. The IVP will often show lesions at the tip of multiple calcynes in both kidneys. The most sensitive tests are a retrograde IVP and urteroscopy, which are reserved for patients with problems requiring intervention to reduce bleeding.

When patients with sickle cell trait present with substantial bleeding the standard practice in the past was to place patients at bed rest under sedation in hospital, and give intra-venous hydration, reduce tonicity by infusion of water, alkalinize the urine by giving sodium bicarbonate, and administer diuretics to sustain a high urine output. These measures have been justified by theoretical considerations rather than substantial controlled observations (6, 31).

A subsequent controlled trial of aggressive reduction in serum tonicity was conducted for patients with sickle cell disease, using hypotonic fluids and infusions of DDAVP. A clear effect reducing in vivo hemolysis required a dangerous level of hyponatremia. This observation dampened enthusiasm for aggressive methods to reduce serum tonicity in sickle cell trait. With no controlled trials this treatment has now fallen out of use. Physicians at our institution seldom put patients on complete bed rest when not hospitalized but ask people with persistent hematuria to avoid exercise while continuing to perform sedentary work. Alkalinization of urine has been considered useful, but controlled studies have not been done. Patients are often given 800 to 1,000 meq per day, in the hope that less polymer will form with a lower urine pH. The lower urine pH is thought to encourage the passage of pieces of renal tissue which may slough from the papillae during gross bleeding.

With more experience treating sickle cell disease all of these measures have been pursued less completely. Most physicians place patients on restricted activity, continuing sedentary office work without exercise, and omit sedation. Water infusion is not used. Patients are hydrated with half normal saline to sustain a high urine output and keep serum osmolality in the low normal range. Sodium bicarbonate 650 to 1200mg per day is sometimes used based on anecdotal experience. This might assist in excretion of pieces of necrotic renal papillae which may slough into the calyces during hematuria. Some physicians give modest doses of furosemide supported by adequate intravenous hydration to sustain a high urine output.

If these measures do not work in a few days, it is now customary to use antifibrinolytic agent, usually epsilon aminocaproic acid (EACA) (32). These agents inhibit urokinase, the major fibrinolytic enzyme in the urinary tract. They are concentrated in the urine, so the doses required to reduce hematuria may be 75-fold less than those required to inhibit serum fibrinolysis. They are used at doses far below those producing a systemic coagulopathy. The main side effect is an increased risk of clot formation in the urinary collecting system. Two leading investigators in this field stated the following: "until 1964 nephrectomy was needed in 50% of those patients with severe persistent hemorrhage. In that year Immergut and Stevenson showed the favorable effect of EACA in the control of hematuria associated with hemoglobinopathies." (6). They reported four consecutive cases of hematuria with sickle cell trait or disease, who responded to EACA (33). Since the use of EACA, nephrectomy for intractable hematuria has become quite rare.

An important controlled study of EACA was conducted a decade later (34). The duration of hematuria was examined for patients with hemoglobin S. Nineteen episodes treated with EACA stopped bleeding at a mean of 2.2 0.3 days versus 11 episodes untreated with EACA which lasted 4.5 1.9 days. The treated patients were selected for a more severe prior history (bleeding for 22.5 6 days prior to EACA versus controls bleeding for 10.3 1,7 days prior to observation). This treatment utilized 6 to 8 grams EACA per day in four to six divided doses. The authors imply that therapy should continue at least three days beyond clearing of gross hematuria but they do not state the duration used. There was a trend toward shorter hematuria with an additional loading dose of five grams of EACA at the start of therapy but the difference in time to recovery was not statistically significant. This study reported a high incidence of ureteral obstruction by clots accompanied by flank pain (15/38 episodes with an IVP) which resolved without specific therapy over 2 to 37 days. Surprisingly, urteral obstruction by clot was not more frequent with than without EACA. Five patients received EACA while clot was present, three with pre-treatment clots and two with clots starting during EACA. These patients cleared their ureteral clots despite completing the planned course of EACA. In this small study preexisting or new clots in the ureters were not considered an indication to avoid using EACA at the low doses stated.

Formal controlled studies to establish the best dose range and duration of EACA for hematuria associated with hemoglobin S are incomplete (34). An uncontrolled study reported the use of loading doses of EACA, 5 grams rapidly and then 10 grams over 12 hours, repeated once if hematuria continued (quoted in 34). After cessation of hematuria they kept the patients in hospital and gave a tapering dose for three days , starting with EACA at 15 grams orally per day and continuing with the lowest dose for another three days as an outpatient. This was successful for 8/11 patients with sickle cell trait and hematuria.

A number of clinicians performed controlled studies of EACA for prostatectomy during the 1960's and 1970's. The now outmoded technique for prostatectomy in use at that time resulted in substantial prolonged hematuria which was managed with EACA. These studies are most valuable for documenting tolerated doses of EACA and risk of complications. Probably EACA was being used at higher doses and longer durations than can be justified by studies of hematuria with sickle cell trait. Controlled studies demonstrated no increase in thromboembolism with administration of 6 grams of EACA in one day or 18 grams in 18 hours.

The largest published experience using EACA to treat hematuria was a report from Scandinavia, in which 526 patients with various forms of hematuria were treated (35). These authors felt that EACA in doses of 3 grams 3 to 4 times per day did not result in thromboembolism. The most relevant group in this study was a collection of patients with essential hematuria who were treated with 3 grams of EACA three times per day for a period of six weeks after cessation of hematuria. Contrary to claims made by McInnes (31), there are no reports of treatment of hematuria with sickle cell trait in this text. There were no controlled observations to justify the long duration of treatment after cessation of hematuria. The largest single group of patients they studied were patients with hematuria post-prostatectomy (250 patients), many of whom received larger doses of EACA. Interpretation of this post-prostatectomy experience with EACA is difficult because they used heparin at 650 units/hour to decrease risk of thromboembolsim. Several authors have quoted a remark that as little as 2 grams of EACA per day might be adequate to treat post-surgical bleeding disorders (6, 31). Review of the context in which this remark was made shows that it cannot be regarded as more than unsubstantiated opinion.

The best dose and duration for use of EACA in treatment of hematuria related to sickle cell trait has not been adequately investigated. The best documented effective dose schedule is 3 grams three times to four times per day for a period of one week, expecting to stop hematuria in two to three days for most patients. Whether given orally or intravenously seems to make little difference. The optimal treatment duration after macroscopic hematuria has stopped remains unknown with a range of clinical experience from three days to six weeks. Our recommendation is to continue with 9 to 12 grams of EACA daily for one to two weeks after cessation of hematuria using the longer period for patients who seem at high risk for recurrence.

Occasionally bleeding is so brisk or so persistent that it is necessary to perform invasive surgery to visualize bleeding sites to identify the pathology at those sites and to stop the bleeding by local measures in order to save the patient and the kidney. Often the surgeon will find multiple damaged calyces in both kidneys. Bleeding is unilateral for 90% of cases, with the left kidney the dominant site. Removal of necrotic or sloughed tissue may be necessary. Bleeding can be stopped by applying local pressure, by cauterizing small vessels which are bleeding, or by using chemicals thought to promote coagulation of injured surfaces.

It is important to avoid nephrectomy because the patient then acquires a high risk of losing both kidneys. At our hospital approximately 20 individuals with sickle cell trait are admitted for hematuria per year and approximately 5-8 patients have undergone surgery to stop bleeding with consideration of nephrectomy over the past ten years. All of these responded to local measures and avoided nephrectomy. In part because these cases are rare no controlled demonstration of efficacy has been conducted. In the future it may be possible to use new recombinent coagulant proteins under development which promote clotting to stop bleeding by systemic or by local treatments.

Studies in Jamaica, England and America established that the rates of urinary tract infection are higher for women with sickle cell trait in comparison to racially matched controls (1, 2, 4, 6). This is best established for asymptomatic bacteruria of pregnancy, in which the rate is approximately doubled with sickle cell trait (36). Rates of pyelonephritis may be modestly increased during pregnancy. No increase in urinary tract infection was noted among men in the large Veteran's Hospital study (15).

Studies of families with autosomal dominant polycystic kidney disease indicate that the incidence of end stage renal failure from this disorder is identical for whites and blacks, but that age of onset of end stage renal failure is lower for black people with sickle cell trait (38 years versus 48 years, p < 0.003). Half of 12 black patients on dialysis for this disorder had sickle cell trait, as opposed to 7.5% of 80 black patients on renal dialysis for other conditions. Sickle cell trait is an important risk factor for early onset of renal failure in patients with autosomal dominant polycystic kidney disease (37).

The dominant tumor mass, from 4 to 12 cm diameter, was in the renal medulla. Satellite lesions in the renal cortex and pelvic soft tissues and invasions of veins and lymphatics were usually present. "The lesions exhibited a reticular, yolk sac-like, or adenoid cystic appearance, often with poorly differentiated areas in a highly desmoplastic stroma admixed with neutrophils and usually marginated by lymphocytes". In all cases metastases outside the kidney were noted at the time of nephrectomy. The mean survival after surgery was 15 weeks.

Radiologic studies demonstrated tumors arising from the central kidney, growing in an infiltrative pattern and invading the renal sinus. A few cases demonstrated caliectasis without pelviectasis and one case showed tumor necrosis into the collecting system. Contrast enhancement and echotexture were heterogeneous. A single angiogram showed hypovascularity.

Six additional patients were found in military records (39). All were young black adults, aged 24 to 36 years. Average survival from diagnosis was 3 months. Cytogenic abnormalities included monosomy 11 in 4/4 patients and abnormalities of chromosome three. Since 1995 at least seven other case reports have been published. Survival has been poor with minimal response to a wide variety of chemotherapy agents and some immunotherapies.

This very rare carcinoma has unusual biologic features since it is largely restricted to patients of African ancestry who are between 11 and 39 years of age. The relative rates of presentation with sickle cell trait versus sickle cell disease are approximately the same as the prevalence of these two genotypes (40 to one). In contrast to this the prevalence of renal cell carcinoma, a much more common tumor in this age group, is nearly 17 times higher than predicted in people with sickle cell disease but not higher with sickle cell trait (40). Early diagnosis of renal medullary carcinoma at a time which would improve survival has not yet been possible.

In their large study of hospitalized veterans at a median age of 49 years, Heller et al. found a statistically significant association between surrogate markers for pulmonary embolism and sickle cell trait (15). The diagnosis of pulmonary embolism was made for 2.2% of those with sickle cell trait versus 1.5% (95% CI of 1.1 to 1.9). Since pulmonary angiograms were only rarely performed, diagnosis depended upon surrogate markers with a low specificity. Dr. Heller was therefore reluctant to regard these statistically significant results as clinically meaningful and feels that the observation of increased incidence of pulmonary embolism in this population is not adequately substantiated.

Among patients with sickle cell trait diagnosed with pulmonary embolism the frequency of thrombophlebitis was significantly higher but the frequency of hemoptysis was significantly lower. This study demonstrated a two-fold increase in essential hematuria (see above). This large study did not find an association between sickle cell trait and risk of vascular complications of diabetes, pyelonephritis, in-hospital mortality from acute myocardial infarction, or mortality or hospital stay post-surgical hospitalization. The combination of erythrocyte glucoe-6-dehydrogenase deficiency with sickle cell disease had no effect on mortality or length of stay, including the subset of patients with pneumonia. There was no significant decline in the fraction of elderly patients with sickle cell trait in comparison to younger patients, confirming no increased mortality with sickle cell trait.

Isolated case reports of unusual adverse events raise the possibility that surgery involving hypoxia or reduced perfusion could result in vaso-occlusion and serious complications for people with sickle cell trait. Some have recommended exchange transfusion to reduce the fraction of cells containing hemoglobin S prior to the tourniquet surgery (4) or for intra-thoracic surgery, especially open-heart surgery on cardio-pulmonary bypass (41). However, the best published controlled study appeared to show no additional risk for people with sickle cell trait who were not transfused, including some intra-thoracic cases (42). A subsequent controlled study of open heart surgery in Africa was interpreted as showing no adverse effects related to sickling for eleven patients with sickle cell trait and two with doubly heterozygous sickle cell disease (43). However, two patients with sickle cell trait died from complications of surgery. The authors attributed these deaths to unavoidable risk from severe cardiac lesions rather than any effect from sickling. Authorities differ in their recommendations for high risk surgery on patients with sickle cell trait, several favoring no exchange transfusion (2, 44) and others advocating exchange transfusion for both cardiac by-pass surgery and tourniquet surgery (42), or limiting this to tourniquet surgery (41).

A number of studies have shown association of sickle cell trait with prematurity and lower birth weight of babies (1, 2, 4). However, data supporting these trends indicate small effects not seen in all studies. These effects seem to have little real public health implication for the long term outcome for mothers or babies.

People with sickle cell trait are more susceptible to complications following treatment of hyphema. Slow flow of relatively hypoxic fluid in the chamber of the eye out of the filtration apparatus is a location in which both polymerization of hemoglobin S and obstruction of flow by rigid erythrocytes is likely (1, 2, 4). This can result in glaucoma and secondary hemorrhage. In a study from Tennessee of 99 eyes from 97 children with hyphema, secondary hemorrhage only occurred in 14 eyes of 13 children with sickle cell trait. The frequency with sickle cell trait was 64%, significantly higher than among 57 eyes without sickle cell trait (0%).

Complications attributed by some to sickle cell trait include proliferative retinopathy, worsening of diabetic retinopathy, stroke, myocardial infarction, leg ulcers, avascular necrosis and arthritis of joints, and increased frequency of the bends from diving. There is no convincing evidence that sickle cell trait increases the incidence of these problems. Some case reports may represent situations in which other variants of beta (4) or alpha globin produced undiagnosed sickle cell disease (4, 45). Others may be the consequence of phenotypes with increased 2,3-DPG or with arterial desaturation which has increased the rate of polymerization of hemoglobin S sufficiently to convert a patient with sickle cell trait into phenotypic sickle cell disease (4, 42, 46).

A study of 355 hospitalized black men with sickle cell trait was conducted to examine stratification of risk by hemoglobin S fraction for pulmonary embolism, thrombophlebitis, myocardial infarction, stroke, and idiopathic hematuria. Hemoglobin S did not influence the frequency of these syndromes, providing evidence that sickling is not associated with these forms of vascular disease. However, the absence of a significant difference for hematuria, which was influenced by hemoglobin S concentration in a larger study, suggests that this study was not sufficiently sensitive.

A summary of the risks associated with sickle cell trait is as follows.

Follow this link:
Sickle Cell Trait - sickle. bwh .harvard.edu

Posted in Cell Medicine | Comments Off on Sickle Cell Trait – sickle. bwh .harvard.edu

Stem Cell Therapy New York City | Regenerative Medicine …

Posted: November 29, 2016 at 8:44 am

Scientists and doctors have made tremendous advances in moving regenerative medicine into the mainstream as a treatment for many diseases and disorders. Regenerative medicine takes advantage of our natural ability to heal ourselves by using the healthy adult stem cells found throughout the body. Laboratory and clinical research has shown that it is possible to use adult stem cells to restore lost, damaged or aging cells to effectively regenerate tissue and provide some patients with an alternative to surgery. Regenerative therapies are showing promise in orthopedic medicine, wound care, nerve restoration, and a variety of cardiovascular, neuromuscular, and autoimmune conditions.

Adult stem cells were discovered over 40 years ago when researchers found that cells derived from bone marrow had the ability to form various tissues. Adult stem cells are early stage cells that, under the right conditions, are capable of developing into other types of cells and hold the potential to regenerate damaged tissue.

AUTOLOGOUS ADULT STEM CELLS (ASCS) are being used to treat many types of chronic pain and degeneration. Currently doctors are treating shoulder, knee, hip, and spine degeneration, in addition to soft tissue (muscle, tendon, ligament) and other bone related injuries.

The first step is to determine if you are a good candidate for an adult stem cell procedure. Your physician will want a history of your injury and a physical examination along with any x-rays, and an MRI. While stem cell therapy maybe appropriate for certain conditions, it is not applicable for every condition. However, it is has proven to be a viable option for several individuals suffering from pain. Good candidates for adult stem cell treatment usually are:

Every patient is different, the success of the stem cell therapy is dependent on the severity of your condition and your bodys response to stem cell therapy.

Overview of the Procedure

An adult stem cell procedure harnesses and amplifies the bodys natural mechanism for healing and anti-inflammation. Once you have been identified as a good candidate for the procedure, a member of our team will review the procedure with you and answer any questions that you may have. A brief overview of the procedure is below:

Once the procedure is complete, our staff will allow you to rest and will create a customized personal rehabilitation program for recovery. We will either ask you to come back for a few post-operative appointments or follow up with you by phone, email, or mail so we can track you healing progress.

Potential Applications

Frequently Asked Questions (FAQs)

Q: What are adult stem cells?

A: Adult stem cells are unspecialized or undifferentiated cells, capable of two processes: self-renewal and differentiation. They are vital to maintaining tissues in the body such as internal organs, skin, and blood.

Q: What is Regenerative Medicine?

A: Regenerative Medicine is a new and advancing scientific field focused on the repair and regeneration of damaged tissue utilizing stem cells.

Q: What is the difference between adult stem cells and embryonic stem cells?

A: Adult stem cells are found in mature adult tissues including bone marrow and fat, while embryonic stem cells (ESCs) are not found in the adult human body. ESCs are obtained from donated in vitro fertilizations, which raises many ethical concerns. Because ESCs are not autologous, there is a possibility of immune rejection. Adult stem cells do not raise ethical issues nor pose any risks for immune rejection.

Q: Does Dr. Youm use embryonic stem cells in clinical procedures?

A: No, Dr. Youms approach to cell therapy relies only on autologous adult stem cells isolated from the patient during surgery. He does not participate in embryonic stem cell research or use embryonic stem cells in clinical applications.

Q: Are there ethical issues associated with harvesting adult stem cells?

A: No, adult stem cells do not raise ethical questions as they are harvested from the patients body.

Q: Are there cancer-causing risks associated with adult stem cell treatments?

A: No. Where embryonic stem cells have been shown to form teratomas (germ cell tumors), there is no data that suggests adult stem cells have the same potential to promote the development of tumors.

Q: Where do adult stem cells come from?

A: In adults, stem cells are present within various tissues and organ systems, the most common being bone marrow and adipose or fat tissues. Other sources include the liver, epidermis, retina, skeletal muscle, intestine, brain, placenta, umbilical cord and dental pulp.

Q: How does Dr. Youm obtain adult stem cells for use in cell treatment?

A: Dr. Youm currently has a system that uses adult stem cells from bone marrow and these stem cells are obtained through aspiration during your procedure.

Q: How are adult stem cells used in surgical procedures?

A: Adult stem cells are used to treat patients with damaged tissues due to age or deterioration. During a procedure, stem cells are isolated from the patient, concentrated and delivered back to the site of injury to assist in the healing process.

Q: Are there different types of adult stem cells?

A: Yes, there are many types of adult stem cells found in the body, which have variable differentiation potentials. The adult stem cells that aid in the repair of damages tissue are multipotent, mesenchymal stem cells. These are located in bone marrow and adipose (fat) tissue.

Q: Are the harvested adult stem cells expanded in a laboratory setting prior to delivery back to the patient?

A: No, Dr. Youm does not use in vitro expansion. The cells are harvested, processed in the operating room and delivered back to the patient at point of care.

Q: How do stem cells know what type of tissue to develop into?

A: The differentiation of stem cells is dependent on many factors, including cell signaling and micro-environmental signals. Based on these cues, stem cells are able to develop into healthy tissue needed to repair damaged tissue. For example, multipotent stem cells delivered to damaged bone will develop into bone cells to aid in tissue repair. The exact mechanism of lineage-specific differentiation is unknown at this point.

Q: Will my body reject the stem cells?

A: No, adult stem cells are autologous and non-immunogenic.

Q: Is stem cell therapy safe?

A: Yes, and ask your doctor what clinical studies have been done to show that stem cells are safe and effective.

Q: Where are stem cells currently being used?

A: Stem cells are currently being used in both laboratory and clinical settings. Laboratories are using human and animal derived stem cells to conduct in vitro studies as well as in vivo studies with small and large animals. Autologous adult stem cells are currently being used in hospitals and clinics during surgery to aid in the repair of damaged tissues.

Q: How long will the stem cells last?

A: It will depend on your injury, the area that is treated and your response to the therapy.

Q: What is the recovery like after a stem cell procedure?

A: If you have a joint injection, you typically can go back to work. It is advised to limit load bearing activities for at least 2 weeks. If you had disc injections, you should take it easy for a few days. Non-steriodal, anti-inflammatory medications (NSAIDS) should be withheld for 72 hours pre-procedure and one week after the procedure.

Q: What is the difference between autologous and allogeneic cells?

A: Autologous cells are taken from the same patient, typically at point-of-care. Allogeneic cells are taken from another patient and are often manipulated before they are given to another patient.

Q: Why use adult stem cell therapy rather than pharmaceuticals or genetic treatments?

A: Adult stem cells are from the body and generate natural proteins and therapeutic biochemicals, decrease inflammation, are anti-bacterial, and recruit other cells to heal the injured site. Pharmaceutical treatments only provide drugs with minimally effective dosages that may cause unwanted side effects. Over dosage can be dangerously toxic or even carcinogenic. Genetic therapy is still unproven and serious concerns exist about it causing cancer due to genetically manipulated cells.

Q: What is the difference between autologous and allogeneic cells?

A: Autologous cells are taken from the same patient, typically at point-of-care. Allogeneic cells are taken from another patient and are often manipulated.

Q: How much will it cost?

A: Most insurance will not cover stem cell procedures. Ask your doctor for payment options

The use of Stem cells in Hip Therapies

The use of Stem cells in Knee Therapies

The use of Stem cells in Shoulder Therapies

You will need the Adobe Reader to view and print these documents.

Visit link:
Stem Cell Therapy New York City | Regenerative Medicine ...

Posted in New York Stem Cells | Comments Off on Stem Cell Therapy New York City | Regenerative Medicine …

Page 1,706«..1020..1,7051,7061,7071,708..1,7201,730..»