Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Blood ; 119(16): 3712-23, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22282502

ABSTRACT

Adult-type lympho-myeloid hematopoietic progenitors are first generated in the aorta-gonad-mesonephros region between days 27 and 40 of human embryonic development, but an elusive blood forming potential is present earlier in the underlying splanchnopleura. In the present study, we show that angiotensin-converting enzyme (ACE, also known as CD143), a recently identified cell-surface marker of adult human hematopoietic stem cells, is already expressed in all presumptive and developing blood-forming tissues of the human embryo and fetus: para-aortic splanchnopleura, yolk sac, aorta-gonad-mesonephros, liver, and bone marrow (BM). Fetal liver and BM-derived CD34(+)ACE(+) cells, but not CD34(+)ACE(-) cells, are endowed with long-term culture-initiating cell potential and sustain multilineage hematopoietic cell engraftment when transplanted into NOD/SCID mice. Furthermore, from 23-26 days of development, ACE expression characterizes rare CD34(-)CD45(-) cells concentrated in the hemogenic portion of the para-aortic splanchnopleura. ACE(+) cells sorted from the splanchnopleura generated colonies of hematopoietic cells more than 40 times more frequently than ACE(-) cells. These data suggest that, in addition to being a marker of adult human hematopoietic stem cells, ACE identifies embryonic mesodermal precursors responsible for definitive hematopoiesis, and we propose that this enzyme is involved in the regulation of human blood formation.


Subject(s)
Bone Marrow/embryology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Liver/embryology , Peptidyl-Dipeptidase A/metabolism , Animals , Antigens, CD34/metabolism , B-Lymphocytes/cytology , Cell Lineage/physiology , Female , Granulocytes/cytology , Hematopoietic Stem Cell Transplantation , Humans , Killer Cells, Natural/cytology , Leukocyte Common Antigens/metabolism , Liver/cytology , Mice , Mice, Inbred NOD , Mice, SCID , T-Lymphocytes/cytology , Transplantation, Heterologous
2.
J Neuropathol Exp Neurol ; 69(12): 1247-55, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21107137

ABSTRACT

The relative contributions of Alzheimer disease (AD) and vascular lesion burden to the occurrence of cognitive decline are more difficult to define in the oldest-old than they are in younger cohorts. To address this issue, we examined 93 prospectively documented autopsy cases from 90 to 103 years with various degrees of AD lesions, lacunes, and microvascular pathology. Cognitive assessment was performed prospectively using the Clinical Dementia Rating scale. Neuropathologic evaluation included the Braak neurofibrillary tangle (NFT) and ß-amyloid (Aß) protein deposition staging and bilateral semiquantitative assessment of vascular lesions. Statistics included regression models and receiver operating characteristic analyses. Braak NFTs, Aß deposition, and cortical microinfarcts (CMIs) predicted 30% of Clinical Dementia Rating variability and 49% of the presence of dementia. Braak NFT and CMI thresholds yielded 0.82 sensitivity, 0.91 specificity, and 0.84 correct classification rates for dementia. Using these threshold values, we could distinguish 3 groups of demented cases and propose criteria for neuropathologic definition of mixed dementia, pure vascular dementia, and AD in very old age. Braak NFT staging and severity of CMI allow for defining most of demented cases in the oldest-old. Most importantly, single cutoff scores for these variables that could be used in the future to formulate neuropathologic criteria for mixed dementia in this age group were identified.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Dementia, Vascular/complications , Dementia, Vascular/pathology , Age Factors , Aged, 80 and over , Alzheimer Disease/psychology , Cohort Studies , Dementia, Vascular/psychology , Female , Humans , Male , Prospective Studies
3.
Int J Dev Biol ; 54(6-7): 1061-5, 2010.
Article in English | MEDLINE | ID: mdl-20711983

ABSTRACT

Hematopoietic stem cells (HSC) are at the origin of the adult hematopoietic system. They give rise to all blood cells through a complex series of proliferation and differentiation events that occur throughout the lifespan of the individual. Because of their potential clinical importance in transplantation, recent research has focused on the developmental origins of embryonic HSC. During development in vertebrate embryos, two independent anatomical sites generate hematopoietic cells. The yolk sac is responsible for a first ephemeral hematopoiesis, characterized by the early appearance of hematopoietic progenitors with limited development ability that rapidly differentiate toward erythro-myeloid lineages. Self-renewing, multipotent adult-type HSC that also exhibit B and T lymphoid potentials emerge autonomously in the aorta/gonad/mesonephros (AGM) region inside the embryo. In this review, we provide a brief summary of recent developments regarding the origins of hematopoietic stem cells in the early human embryo. The recent discovery that angiotensin-converting enzyme (ACE) is a novel cell surface marker of human HSC is discussed in detail.


Subject(s)
Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic System/embryology , Cell Lineage , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Peptidyl-Dipeptidase A/metabolism
4.
Acta Vet Hung ; 57(2): 239-46, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19584037

ABSTRACT

This paper introduces two novel monoclonal antibodies, designated GTr1 and GTr2, which recognise guinea fowl thrombocyte surface antigen(s). The antibodies were tested in embryos and adult birds. GTr1 and GTr2 staining emerged at embryonic days 12 and 7, respectively. After embryonic day 12 there was no difference in staining pattern between the two monoclonal antibodies. The isotype of the antibodies is IgG1. The antibodies did not react with any other haematopoietic cells of guinea fowl, and there was no species cross-reaction with chicken, turkey and quail. The antibodies can be used in interspecies chimeric and parabiotic experiments to identify cells of guinea fowl origin.


Subject(s)
Antibodies, Monoclonal/immunology , Blood Platelets/immunology , Galliformes/blood , Animals , Antigens, Surface , Blood Platelets/cytology , Chickens/immunology , Quail/immunology , Turkeys/immunology
5.
Blood ; 111(8): 4055-63, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-17993616

ABSTRACT

Previous studies revealed that mAb BB9 reacts with a subset of CD34(+) human BM cells with hematopoietic stem cell (HSC) characteristics. Here we map BB9 expression throughout hematopoietic development and show that the earliest definitive HSCs that arise at the ventral wall of the aorta and surrounding endothelial cells are BB9(+). Thereafter, BB9 is expressed by primitive hematopoietic cells in fetal liver and in umbilical cord blood (UCB). BB9(+)CD34(+) UCB cells transplanted into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice contribute 10-fold higher numbers of multilineage blood cells than their CD34(+)BB9(-) counterparts and contain a significantly higher incidence of SCID-repopulating cells than the unfractionated CD34(+) population. Protein microsequencing of the 160-kDa band corresponding to the BB9 protein established its identity as that of somatic angiotensin-converting enzyme (ACE). Although the role of ACE on human HSCs remains to be determined, these studies designate ACE as a hitherto unrecognized marker of human HSCs throughout hematopoietic ontogeny and adulthood.


Subject(s)
Fetus/enzymology , Hematopoietic Stem Cells/enzymology , Hematopoietic System/enzymology , Peptidyl-Dipeptidase A/metabolism , Adult , Animals , Antibodies, Monoclonal , Antibody Specificity/drug effects , Antigens, CD34/metabolism , Cell Count , Cell Lineage/drug effects , Cell Proliferation/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/enzymology , Female , Fetus/drug effects , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic System/embryology , Humans , Lisinopril/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects
6.
Ann N Y Acad Sci ; 1106: 223-32, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17360801

ABSTRACT

Human hematopoiesis proceeds transiently in the extraembryonic yolk sac and embryonic, then fetal liver before being stabilized in the bone marrow during the third month of gestation. In addition to this classic developmental sequence, we have previously shown that the aorta-gonad-mesonephros (AGM) embryonic territory produces stem cells for definitive hematopoiesis from 27 to 40 days of human development, through an intermediate blood-forming endothelium stage. These studies have relied on the use of traditional markers of human hematopoietic and endothelial cells. In addition, we have recently identified and characterized a novel surface molecule, BB9, which typifies the earliest founders of the human angiohematopoietic system. BB9, which was initially identified with a monoclonal antibody raised to Stro-1(+) bone marrow stromal cells, recognizes in the adult the most primitive Thy-1(+) CD133(+) Lin(-), non-obese diabetic--severe combined immunodeficiency disease (NOD-SCID) mouse engrating hematopoietic stem cells (HSCs). In the 3- to 4-week embryo, BB9 expression typifies a subset of splanchnopleural mesodermal cells that migrate dorsally and colonize the ventral aspect of the aorta where they establish a population of hemogenic endothelial cells. We have indeed confirmed that hematopoietic potential in the human embryo, as assessed by long-term culture-initiating cell (LTC-IC) and SCID mouse reconstituting cell (SRC) activities, is confined to BB9-expressing cells. We have further validated these results in the model of human embryonic stem cells (hESCs) in which we have modeled, through the development of hematopoietic embryoid bodies (EBs), primitive and definitive hematopoieses. In this setting, we have documented the emergence of BB9(+) hemangioblast-like clonogenic angiohematopoietic progenitors that currently represent the earliest known founders of the human vascular and blood systems.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Neovascularization, Physiologic , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Cell Movement , Endothelial Cells/cytology , Glycoproteins/biosynthesis , Hematopoiesis , Humans , Liver/embryology , Mice , Mice, Inbred NOD , Mice, SCID , Models, Biological , Peptides
SELECTION OF CITATIONS
SEARCH DETAIL
...