Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Racial Ethn Health Disparities ; 9(5): 1626-1631, 2022 10.
Article in English | MEDLINE | ID: mdl-34286478

ABSTRACT

Racism impacts every aspect of medicine, including the careers and lives of Black physicians. The story of William Augustus Hinton (1883-1959), who invented the Hinton Test for syphilis before becoming the first African American full professor at Harvard University in 1949, offers an instructive perspective on the intersection of interpersonal and systemic racism, and personal determination, just over our historical horizon. Yet there are sobering and instructive lessons throughout this history. Hinton had to navigate prejudice throughout his career. Indeed, while there is much to be inspired by in the telling of Hinton's story, the forms of racism faced by Hinton and his contemporaries remain persisting features of academic medicine. This article focuses on encounters with racism that affect the course of medical careers and scientific innovation. Hinton's story holds important implications for many health professionals in the twenty-first century and provides unique insights into the history and impact of interpersonal and systemic racism alike in academic medicine.


Subject(s)
Physicians , Racism , Black or African American/history , Black People , Humans , Racism/history
3.
Nature ; 525(7570): 469-78, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26399828

ABSTRACT

Stem cells self-renew and generate specialized progeny through differentiation, but vary in the range of cells and tissues they generate, a property called developmental potency. Pluripotent stem cells produce all cells of an organism, while multipotent or unipotent stem cells regenerate only specific lineages or tissues. Defining stem-cell potency relies upon functional assays and diagnostic transcriptional, epigenetic and metabolic states. Here we describe functional and molecular hallmarks of pluripotent stem cells, propose a checklist for their evaluation, and illustrate how forensic genomics can validate their provenance.


Subject(s)
Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genomics , Humans
4.
Stem Cell Reports ; 1(1): 5-17, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-24052937

ABSTRACT

Cellular reprogramming was recently "crowned" with the award of the Nobel Prize to two of its groundbreaking researchers, Sir John Gurdon and Shinya Yamanaka. The recent link between reprogramming and stem cells makes this appear almost a new field of research, but its historical roots have actually spanned more than a century. Here, the Nobel Prize in Physiology or Medicine 2012 is placed in its historical context.


Subject(s)
Cellular Reprogramming , Nobel Prize , Stem Cell Research/history , Animals , History, 20th Century , History, 21st Century , Humans
5.
Cell Stem Cell ; 12(6): 727-36, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23602541

ABSTRACT

Shwachman-Diamond syndrome (SDS), a rare autosomal-recessive disorder characterized by exocrine pancreatic insufficiency and hematopoietic dysfunction, is caused by mutations in the Shwachman-Bodian-Diamond syndrome (SBDS) gene. We created human pluripotent stem cell models of SDS through knockdown of SBDS in human embryonic stem cells (hESCs) and generation of induced pluripotent stem cell (iPSC) lines from two patients with SDS. SBDS-deficient hESCs and iPSCs manifest deficits in exocrine pancreatic and hematopoietic differentiation in vitro, enhanced apoptosis, and elevated protease levels in culture supernatants, which could be reversed by restoring SBDS protein expression through transgene rescue or by supplementing culture media with protease inhibitors. Protease-mediated autodigestion provides a mechanistic link between the pancreatic and hematopoietic phenotypes in SDS, highlighting the utility of hESCs and iPSCs in obtaining novel insights into human disease.


Subject(s)
Bone Marrow Diseases/pathology , Bone Marrow Diseases/physiopathology , Exocrine Pancreatic Insufficiency/pathology , Exocrine Pancreatic Insufficiency/physiopathology , Induced Pluripotent Stem Cells/pathology , Lipomatosis/pathology , Lipomatosis/physiopathology , Pancreas/pathology , Pancreas/physiopathology , Bone Marrow Diseases/enzymology , Cell Differentiation , Cells, Cultured , Exocrine Pancreatic Insufficiency/enzymology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/enzymology , Lipomatosis/enzymology , Models, Biological , Pancreas/enzymology , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Shwachman-Diamond Syndrome
6.
Proc Natl Acad Sci U S A ; 110(2): E141-50, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23236128

ABSTRACT

During development, the hematopoietic lineage transits through hemogenic endothelium, but the signaling pathways effecting this transition are incompletely characterized. Although the Hedgehog (Hh) pathway is hypothesized to play a role in patterning blood formation, early embryonic lethality of mice lacking Hh signaling precludes such analysis. To determine a role for Hh signaling in patterning of hemogenic endothelium, we assessed the effect of altered Hh signaling in differentiating mouse ES cells, cultured mouse embryos, and developing zebrafish embryos. In differentiating mouse ES cells and mouse yolk sac cultures, addition of Indian Hh ligand increased hematopoietic progenitors, whereas chemical inhibition of Hh signaling reduced hematopoietic progenitors without affecting primitive streak mesoderm formation. In the setting of Hh inhibition, induction of either Notch signaling or overexpression of Stem cell leukemia (Scl)/T-cell acute lymphocytic leukemia protein 1 rescued hemogenic vascular-endothelial cadherin(+) cells and hematopoietic progenitor formation. Together, our results reveal that Scl overexpression is sufficient to rescue the developmental defects caused by blocking the Hh and Notch pathways, and inform our understanding of the embryonic endothelial-to-hematopoietic transition.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Endothelium/physiology , Hedgehog Proteins/metabolism , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Colony-Forming Units Assay , Embryo, Mammalian , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Hematopoietic Stem Cells/physiology , In Situ Hybridization , Mice , Mice, Inbred C57BL , T-Cell Acute Lymphocytic Leukemia Protein 1 , Zebrafish
7.
Stem Cell Rev Rep ; 8(2): 551-60, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22278132

ABSTRACT

Easily accessed via venipuncture, blood has been an object of study for centuries. Direct sampling of the hematopoietic tissues in the bone marrow also presents a rather low bar for biopsy acquisition from living donors, especially when compared to other systems in the body such as the heart or brain. This relatively straight-forward ability to obtain cells from the primary anatomical locations of blood cell genesis and differentiation combines with a reliable transplantation assay and well-described surface markers to make the hematopoietic stem cell (HSC) the best understood of all tissue stem cells. HSC biology has been extensively though incompletely investigated over the years. The field continually refreshes itself as new findings require us to reevaluate our understanding of hematopoiesis. After providing a brief overview of the hematopoietic system in general, this review will touch on recent findings in three areas: (1) the niche, (2) HSC migration, and (3) challenges to the "classical" model of hematopoietic ontogeny.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Models, Biological , Cell Lineage , Cell Movement , Genetic Therapy , Humans , Stem Cell Niche
8.
Nat Biotechnol ; 29(12): 1117-9, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22119740

ABSTRACT

We compared bona fide human induced pluripotent stem cells (iPSCs) derived from umbilical cord blood (CB) cells and neonatal keratinocytes (K). As a consequence of both incomplete erasure of tissue-specific methylation and aberrant de novo methylation, CB-iPSCs and K-iPSCs were distinct in genome-wide DNA methylation profiles and differentiation potential. Extended passage of some iPSC clones in culture did not improve their epigenetic resemblance to embryonic stem cells, implying that some human iPSCs retain a residual 'epigenetic memory' of their tissue of origin.


Subject(s)
Cell Differentiation , Cell Lineage , DNA Methylation , Epigenesis, Genetic , Fetal Blood/metabolism , Induced Pluripotent Stem Cells/metabolism , Keratinocytes/metabolism , Fetal Blood/cytology , Gene Expression Regulation , Genome, Human , Humans , Induced Pluripotent Stem Cells/cytology , Keratinocytes/cytology , Microarray Analysis
9.
Genome Med ; 3(7): 44, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21745416
10.
Nat Rev Genet ; 12(4): 266-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21386866

ABSTRACT

Human genetic studies have revealed the molecular basis of countless monogenic diseases but have been less successful in associating phenotype to genotype in complex multigenic conditions. Pluripotent stem cells (PSCs), which can differentiate into any cell type, offer promise for defining the functional effects of genetic variation. Here, we recount the advantages and practical limitations of coupling PSCs to genome-wide analyses to probe complex genetics and discuss the ability to investigate epigenetic contributions to disease states. We also describe new ways of using mice and mouse embryonic stem cells (ESCs) in tandem with human stem cells to further define genotype-phenotype relationships.


Subject(s)
Cell Differentiation , Disease/genetics , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Embryonic Stem Cells/metabolism , Epigenomics , Genome , Genotype , Humans , Mice , Phenotype , Pluripotent Stem Cells/metabolism
13.
Blood ; 115(17): 3453-62, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20089964

ABSTRACT

Fanconi anemia (FA) is a genetically heterogeneous, autosomal recessive disorder characterized by pediatric bone marrow failure and congenital anomalies. The effect of FA gene deficiency on hematopoietic development in utero remains poorly described as mouse models of FA do not develop hematopoietic failure and such studies cannot be performed on patients. We have created a human-specific in vitro system to study early hematopoietic development in FA using a lentiviral RNA interference (RNAi) strategy in human embryonic stem cells (hESCs). We show that knockdown of FANCA and FANCD2 in hESCs leads to a reduction in hematopoietic fates and progenitor numbers that can be rescued by FA gene complementation. Our data indicate that hematopoiesis is impaired in FA from the earliest stages of development, suggesting that deficiencies in embryonic hematopoiesis may underlie the progression to bone marrow failure in FA. This work illustrates how hESCs can provide unique insights into human development and further our understanding of genetic disease.


Subject(s)
Embryonic Stem Cells/metabolism , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia Complementation Group D2 Protein , Fanconi Anemia/metabolism , Gene Knockdown Techniques , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line , Embryonic Stem Cells/pathology , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Models, Biological , RNA Interference
14.
Nature ; 459(7250): 1126-30, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19458618

ABSTRACT

The incidence of many cancer types is significantly reduced in individuals with Down's syndrome, and it is thought that this broad cancer protection is conferred by the increased expression of one or more of the 231 supernumerary genes on the extra copy of chromosome 21. One such gene is Down's syndrome candidate region-1 (DSCR1, also known as RCAN1), which encodes a protein that suppresses vascular endothelial growth factor (VEGF)-mediated angiogenic signalling by the calcineurin pathway. Here we show that DSCR1 is increased in Down's syndrome tissues and in a mouse model of Down's syndrome. Furthermore, we show that the modest increase in expression afforded by a single extra transgenic copy of Dscr1 is sufficient to confer significant suppression of tumour growth in mice, and that such resistance is a consequence of a deficit in tumour angiogenesis arising from suppression of the calcineurin pathway. We also provide evidence that attenuation of calcineurin activity by DSCR1, together with another chromosome 21 gene Dyrk1a, may be sufficient to markedly diminish angiogenesis. These data provide a mechanism for the reduced cancer incidence in Down's syndrome and identify the calcineurin signalling pathway, and its regulators DSCR1 and DYRK1A, as potential therapeutic targets in cancers arising in all individuals.


Subject(s)
Down Syndrome/genetics , Inositol/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Animals , Calcineurin/metabolism , Calcium-Binding Proteins , Catechols , Cells, Cultured , DNA-Binding Proteins , Disease Models, Animal , Down Syndrome/metabolism , Endothelial Cells/metabolism , Gene Dosage/genetics , Humans , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Dyrk Kinases
15.
Nature ; 459(7250): 1131-5, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19440194

ABSTRACT

Biomechanical forces are emerging as critical regulators of embryogenesis, particularly in the developing cardiovascular system. After initiation of the heartbeat in vertebrates, cells lining the ventral aspect of the dorsal aorta, the placental vessels, and the umbilical and vitelline arteries initiate expression of the transcription factor Runx1 (refs 3-5), a master regulator of haematopoiesis, and give rise to haematopoietic cells. It remains unknown whether the biomechanical forces imposed on the vascular wall at this developmental stage act as a determinant of haematopoietic potential. Here, using mouse embryonic stem cells differentiated in vitro, we show that fluid shear stress increases the expression of Runx1 in CD41(+)c-Kit(+) haematopoietic progenitor cells, concomitantly augmenting their haematopoietic colony-forming potential. Moreover, we find that shear stress increases haematopoietic colony-forming potential and expression of haematopoietic markers in the para-aortic splanchnopleura/aorta-gonads-mesonephros of mouse embryos and that abrogation of nitric oxide, a mediator of shear-stress-induced signalling, compromises haematopoietic potential in vitro and in vivo. Collectively, these data reveal a critical role for biomechanical forces in haematopoietic development.


Subject(s)
Cell Differentiation , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Stress, Mechanical , Animals , Aorta/cytology , Aorta/embryology , Cell Line , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Embryonic Stem Cells , Endothelium-Dependent Relaxing Factors/pharmacology , Female , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/drug effects , Mice , Nitric Oxide/pharmacology , Pregnancy
16.
Br Med Bull ; 90: 19-35, 2009.
Article in English | MEDLINE | ID: mdl-19376801

ABSTRACT

INTRODUCTION: Cellular reprogramming is the process of directing mature cells to a primitive state of gene expression. SOURCES OF DATA: Medline searches using the keywords 'pluripotency', 'induce' (and derivatives), and/or 'stem' limited to the years 2006 to the present and other selected literature known to the author. AREAS OF AGREEMENT: Since 2006, there has been a cavalcade of scientific works describing so-called 'direct reprogramming' wherein somatic cells are forced into a state of gene expression very similar to embryonic stem cells. These findings build upon prior research using nuclear transfer (cloning) and even older efforts to understand developmental processes. AREAS OF CONTROVERSY: While already of tremendous research value, it remains to be seen how (if) direct reprogramming methodologies will be refined for clinical use. AREAS TIMELY FOR DEVELOPING RESEARCH: A greater understanding of epigenetics, the process by which different patterns of gene expression are established, maintained and redirected, will continue to be enlightened by advances in cellular reprogramming.


Subject(s)
Biomedical Research/trends , Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Epigenesis, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Pluripotent Stem Cells/cytology , Humans
18.
Cell ; 134(5): 877-86, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18691744

ABSTRACT

Tissue culture of immortal cell strains from diseased patients is an invaluable resource for medical research but is largely limited to tumor cell lines or transformed derivatives of native tissues. Here we describe the generation of induced pluripotent stem (iPS) cells from patients with a variety of genetic diseases with either Mendelian or complex inheritance; these diseases include adenosine deaminase deficiency-related severe combined immunodeficiency (ADA-SCID), Shwachman-Bodian-Diamond syndrome (SBDS), Gaucher disease (GD) type III, Duchenne (DMD) and Becker muscular dystrophy (BMD), Parkinson disease (PD), Huntington disease (HD), juvenile-onset, type 1 diabetes mellitus (JDM), Down syndrome (DS)/trisomy 21, and the carrier state of Lesch-Nyhan syndrome. Such disease-specific stem cells offer an unprecedented opportunity to recapitulate both normal and pathologic human tissue formation in vitro, thereby enabling disease investigation and drug development.


Subject(s)
Cell Line , Genetic Diseases, Inborn/pathology , Pluripotent Stem Cells/cytology , Bone Marrow Cells/cytology , Fibroblasts/cytology , Humans , Karyotyping , Mesenchymal Stem Cells/cytology , Mutation
19.
South Med J ; 101(1): 79-82, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18176297

ABSTRACT

Recent research points to the future of regenerative medicine. In the past year, a handful of research groups have demonstrated that mature, adult cells could be "reprogrammed" to a very primitive, embryonic state via the forced expression of four genes (Oct-3/4, c-Myc, Klf4, and Sox2). These induced pluripotent cells (or iPS) share features with embryonic stem (ES) cells and generate tissues from all three embryonic germ layers (ectoderm, mesoderm, and endoderm). iPS cells are also capable of the ultimate demonstration of developmental potency, ie, when injected into an early mouse embryo, they contribute to the formation of an entire mouse including its germline. While the reprogramming of human fibroblasts into iPS cells remains to be seen, it is nevertheless difficult to overstate the value that this new research contributes to the field of regenerative medicine and its academic relative developmental biology. Herein, we attempt to bring these monumental works into greater focus and comment on how they work to shape the future of cellular therapies.


Subject(s)
Pluripotent Stem Cells/transplantation , Cell Lineage , Cell- and Tissue-Based Therapy/history , Epigenesis, Genetic , Forecasting , History, 19th Century , History, 20th Century , Humans , Kruppel-Like Factor 4 , Stem Cell Transplantation/trends
20.
Nature ; 451(7175): 141-6, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18157115

ABSTRACT

Pluripotency pertains to the cells of early embryos that can generate all of the tissues in the organism. Embryonic stem cells are embryo-derived cell lines that retain pluripotency and represent invaluable tools for research into the mechanisms of tissue formation. Recently, murine fibroblasts have been reprogrammed directly to pluripotency by ectopic expression of four transcription factors (Oct4, Sox2, Klf4 and Myc) to yield induced pluripotent stem (iPS) cells. Using these same factors, we have derived iPS cells from fetal, neonatal and adult human primary cells, including dermal fibroblasts isolated from a skin biopsy of a healthy research subject. Human iPS cells resemble embryonic stem cells in morphology and gene expression and in the capacity to form teratomas in immune-deficient mice. These data demonstrate that defined factors can reprogramme human cells to pluripotency, and establish a method whereby patient-specific cells might be established in culture.


Subject(s)
HMGB Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Adult , Animals , Cell Differentiation , Cell Shape , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fetus/cytology , Fibroblasts/cytology , Gene Expression Profiling , HMGB Proteins/genetics , Homeodomain Proteins/genetics , Humans , Infant, Newborn , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/transplantation , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors , Teratoma/pathology , Transcription Factors/genetics , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...