Emergence of muscle and neural hematopoiesis in humans

Slides:



Advertisements
Similar presentations
by Ayten Kandilci, and Gerard C. Grosveld
Advertisements

Hematopoietic stem cells: concepts, definitions, and the new reality
Protein kinase B (PKB/c-akt) regulates homing of hematopoietic progenitors through modulation of their adhesive and migratory properties by Miranda Buitenhuis,
Lymphoid precursors are directed to produce dendritic cells as a result of TLR9 ligation during herpes infection by Robert S. Welner, Rosana Pelayo, Yoshinori.
HOXA9 promotes hematopoietic commitment of human embryonic stem cells
by Shawn W. Cochrane, Ying Zhao, Robert S. Welner, and Xiao-Hong Sun
Enhancement of intracellular signaling associated with hematopoietic progenitor cell survival in response to SDF-1/CXCL12 in synergy with other cytokines.
Megakaryocyte Growth and Development Factor-Induced Proliferation and Differentiation Are Regulated by the Mitogen-Activated Protein Kinase Pathway in.
Volume 86, Issue 1, Pages (July 1996)
The Chemokine Receptor CXCR4 Is Required for the Retention of B Lineage and Granulocytic Precursors within the Bone Marrow Microenvironment  Qing Ma,
Human NK cell development in NOD/SCID mice receiving grafts of cord blood CD34+ cells by Christian P. Kalberer, Uwe Siegler, and Aleksandra Wodnar-Filipowicz.
Prospective isolation and global gene expression analysis of the erythrocyte colony-forming unit (CFU-E)‏ by Grzegorz Terszowski, Claudia Waskow, Peter.
Evidence for MPL W515L/K mutations in hematopoietic stem cells in primitive myelofibrosis by Ronan Chaligné, Chloé James, Carole Tonetti, Rodolphe Besancenot,
Bone morphogenetic protein 4 induces efficient hematopoietic differentiation of rhesus monkey embryonic stem cells in vitro by Fei Li, Shijiang Lu, Loyda.
Novel function for interleukin-7 in dendritic cell development
Abnormalities in the myeloid progenitor compartment in Down syndrome fetal liver precede acquisition of GATA1 mutations by Oliver Tunstall-Pedoe, Anindita.
Hyaluronate-Enhanced Hematopoiesis: Two Different Receptors Trigger the Release of Interleukin-1β and Interleukin-6 From Bone Marrow Macrophages by Sophia.
Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients by Devi.
Establishment of Endoderm Progenitors by SOX Transcription Factor Expression in Human Embryonic Stem Cells  Cheryle A. Séguin, Jonathan S. Draper, Andras.
Vascular Endothelial Cells Produce Soluble Factors That Mediate the Recovery of Human Hematopoietic Stem Cells after Radiation Injury  Garrett G. Muramoto,
Human Progenitor Cells Rapidly Mobilized by AMD3100 Repopulate NOD/SCID Mice with Increased Frequency in Comparison to Cells from the Same Donor Mobilized.
Proteasome activity restricts lentiviral gene transfer into hematopoietic stem cells and is down-regulated by cytokines that enhance transduction by Francesca.
The Anemic Friend Virus gp55 Envelope Protein Induces Erythroid Differentiation in Fetal Liver Colony-Forming Units-Erythroid by Stefan N. Constantinescu,
by Anupama Narla, Shilpee Dutt, J
Elucidation of the EP defect in Diamond-Blackfan anemia by characterization and prospective isolation of human EPs by Deena Iskander, Bethan Psaila, Gareth.
Different ploidy levels of megakaryocytes generated from peripheral or cord blood CD34+ cells are correlated with different levels of platelet release.
Bifurcated Dendritic Cell Differentiation In Vitro From Murine Lineage Phenotype-Negative c-kit + Bone Marrow Hematopoietic Progenitor Cells by Yi Zhang,
Identification and characterization of 2 types of erythroid progenitors that express GATA-1 at distinct levels by Norio Suzuki, Naruyoshi Suwabe, Osamu.
The p67 laminin receptor identifies human erythroid progenitor and precursor cells and is functionally important for their bone marrow lodgment by Halvard.
MEK kinase 1 activity is required for definitive erythropoiesis in the mouse fetal liver by Barbara Bonnesen, Cathrine Orskov, Susanne Rasmussen, Peter.
Pak2 regulates myeloid-derived suppressor cell development in mice
by Pratima Chaurasia, Dmitriy Berenzon, and Ronald Hoffman
by Suzanne M. Vercauteren, and Heather J. Sutherland
Distinct classes of c-Kit–activating mutations differ in their ability to promote RUNX1-ETO–associated acute myeloid leukemia by Heidi J. Nick, Hyung-Gyoon.
Volume 3, Issue 5, Pages (November 2014)
Lentiviral-mediated RNAi inhibition of Sbds in murine hematopoietic progenitors impairs their hematopoietic potential by Amy S. Rawls, Alyssa D. Gregory,
Cells Isolated from the Epidermis by Hoechst Dye Exclusion, Small Size, and Negative Selection for Hematopoietic Markers Can Generate B Lymphocyte Precursors 
Cytotoxic CD8+ T Cells Stimulate Hematopoietic Progenitors by Promoting Cytokine Release from Bone Marrow Mesenchymal Stromal Cells  Christian M. Schürch,
Volume 4, Issue 2, Pages (February 2003)
Clinically Relevant Expansion of Hematopoietic Stem Cells with Conserved Function in a Single-Use, Closed-System Bioprocess  Gerard J. Madlambayan, Ian.
Volume 2, Issue 6, Pages (December 2012)
Volume 4, Issue 3, Pages (March 2015)
SHIP is required for a functional hematopoietic stem cell niche
Volume 29, Issue 2, Pages (August 2008)
Volume 43, Issue 2, Pages (August 2015)
Volume 16, Issue 5, Pages (May 2002)
Ravindra Majeti, Christopher Y. Park, Irving L. Weissman 
Volume 10, Issue 6, Pages (December 2004)
David Traver, Koichi Akashi, Irving L. Weissman, Eric Lagasse  Immunity 
Volume 15, Issue 4, Pages (October 2001)
Volume 24, Issue 1, Pages (January 2006)
Twist1 regulates embryonic hematopoietic differentiation through binding to Myb and Gata2 promoter regions by Kasem Kulkeaw, Tomoko Inoue, Tadafumi Iino,
Kiran Batta, Magdalena Florkowska, Valerie Kouskoff, Georges Lacaud 
Volume 3, Issue 6, Pages (December 2014)
Human hematopoietic stem cell maintenance and myeloid cell development in next-generation humanized mouse models by Trisha R. Sippel, Stefan Radtke, Tayla.
Volume 11, Issue 5, Pages (November 2018)
Volume 3, Issue 5, Pages (May 2013)
Human granulocyte-macrophage colony-stimulating factor (hGM-CSF)–dependent in vitro and in vivo proliferation and differentiation of all hematopoietic.
Tumor Necrosis Factor-α- and IL-4-Independent Development of Langerhans Cell-Like Dendritic Cells from M-CSF-Conditioned Precursors  Jean-Baptiste Barbaroux,
SLAM Family Markers Resolve Functionally Distinct Subpopulations of Hematopoietic Stem Cells and Multipotent Progenitors  Hideyuki Oguro, Lei Ding, Sean J.
Reduced numbers and self-renewal of HPCs in the absence of geminin.
Decellularized Wharton jelly matrix: a biomimetic scaffold for ex vivo hematopoietic stem cell culture by Dandan Li, Grace Chiu, Brea Lipe, Richard A.
IL-33, IL-25, and TSLP induce a distinct phenotypic and activation profile in human type 2 innate lymphoid cells by Ana Camelo, Guglielmo Rosignoli, Yoichiro.
A Role for G-CSF Receptor Signaling in the Regulation of Hematopoietic Cell Function but Not Lineage Commitment or Differentiation  Craig L Semerad, Jennifer.
An Mll-Dependent Hox Program Drives Hematopoietic Progenitor Expansion
Volume 17, Issue 2, Pages (August 2002)
Volume 86, Issue 1, Pages (July 1996)
Volume 21, Issue 1, Pages (July 2004)
Α4 Integrins Regulate the Proliferation/Differentiation Balance of Multilineage Hematopoietic Progenitors In Vivo  Alicia G Arroyo, Joy T Yang, Helen.
Granulocyte colony-stimulating factor mobilizes dormant hematopoietic stem cells without proliferation in mice by Jeffrey M. Bernitz, Michael G. Daniel,
Presentation transcript:

Emergence of muscle and neural hematopoiesis in humans by Karen E. Jay, Lisa Gallacher, and Mickie Bhatia Blood Volume 100(9):3193-3202 November 1, 2002 ©2002 by American Society of Hematology

Flow cytometric analysis of cell surface phenotype of cells derived from human muscle and neural tissues.Flow cytometry was used to evaluate the cell surface expression of the human-specific proteins associated with hematopoietic tissue: AC133, and cell dif... Flow cytometric analysis of cell surface phenotype of cells derived from human muscle and neural tissues.Flow cytometry was used to evaluate the cell surface expression of the human-specific proteins associated with hematopoietic tissue: AC133, and cell differentiation markers CD34 and CD45. A representative FACS analysis of de novo–isolated muscle (A) and neural (B) cells is shown, and the percentage of subpopulations expressing single or coexpressing human hematopoietic markers is shown as the average mean ± SEM (n = 4) in each quadrant. Specificity of hematopoietic antibody staining and background signal was determined by comparing muscle and neural cells stained with mouse IgG1 (Ai and Bi) to establish positive quadrant levels to omit fluorescence because of nonspecific binding and auto fluorescence properties of the cells. Results are based on data from 4 independent samples of muscle and neural tissues analyzed in duplicate. Karen E. Jay et al. Blood 2002;100:3193-3202 ©2002 by American Society of Hematology

Human chimerism in the tissue of immune-deficient mice that received intravenous transplants of human fetal tissues.Comparison of human hematopoietic chimerism from hematopoietic and nonhematopoietic sources. Human chimerism in the tissue of immune-deficient mice that received intravenous transplants of human fetal tissues.Comparison of human hematopoietic chimerism from hematopoietic and nonhematopoietic sources. Scatterplots show FACS analysis of mouse BM stained with the pan-leukocyte marker CD45 for the detection of human hematopoietic cells (gated box) after transplantation with isotype control (A), fetal blood (B), fetal liver (C), fetal BM (D), fetal muscle (E), and fetal neural cells (F). Cell doses ranged between 2 × 106 and 5 × 106 to up to 15 × 106 for muscle and neural transplants (n = 43). Karen E. Jay et al. Blood 2002;100:3193-3202 ©2002 by American Society of Hematology

Immunohistochemical and morphologic analysis of human fetal muscle and neural cells using tissue-specific markers and emergence of functional hematopoietic progenitors.Light microscopy was used to visualize cells comprising human muscle (Ai) and neural (Bi)... Immunohistochemical and morphologic analysis of human fetal muscle and neural cells using tissue-specific markers and emergence of functional hematopoietic progenitors.Light microscopy was used to visualize cells comprising human muscle (Ai) and neural (Bi) tissues, showing morphologic features associated with these tissue types. Human muscle tissue specificity was analyzed by immunohistochemistry for the expression of muscle-specific markers recognizing heavy chain of myosin (Aii) and the nuclear DNA binding factor, myogenin (Aiii), whereas human neural tissue demonstrated expression of neural progenitor-specific marker, nestin (Bii) and MAP-2 (Biii), respectively. A representative panel of multilineage hematopoietic colonies derived from muscle (Aiv) and neural (Biv) tissues cultured in the presence of HGF together with BMP-4 and EPO is shown. Human hematopoietic colony types generated from muscle and neural tissues include erythroid burst-forming unit (BFU-E), CFU-granulocyte (G), -macrophage (M), and tetrapotent mixed colonies (granulocyte, erythroid, macrophage, megakaryocyte [GEMM]). Similar results were obtained from 9 independent samples of muscle and neural tissue samples. Magnification × 200. Karen E. Jay et al. Blood 2002;100:3193-3202 ©2002 by American Society of Hematology

Evaluation of human hematopoietic cell fate potential of de novo and cultured muscle and neural cells.Primary human muscle (A) and neural (B) tissues were examined for hematopoietic progenitor capacity (CFU) at isolation (day 0) or after 5 days of in vitro ... Evaluation of human hematopoietic cell fate potential of de novo and cultured muscle and neural cells.Primary human muscle (A) and neural (B) tissues were examined for hematopoietic progenitor capacity (CFU) at isolation (day 0) or after 5 days of in vitro culture in essential media containing 10% CEE, or human hematopoietic growth factors (HGF) alone or with BMP-4 or EPO, as single additions or in combination as indicated. In all culture conditions, media and cytokines were replenished every other day. Single, double, and triple asterisks indicate substantial differences within measured groups of P < .01. Data shown is based on 6 to 13 independent samples analyzed in culture conditions indicated. Karen E. Jay et al. Blood 2002;100:3193-3202 ©2002 by American Society of Hematology

Comparative analysis of human hematopoietic-, muscle-, and neural-derived hematopoietic progenitors.Human tissues indicated were harvested under identical conditions, and functional hematopoietic progenitor capacity was evaluated at day 0 (de novo–isolated ... Comparative analysis of human hematopoietic-, muscle-, and neural-derived hematopoietic progenitors.Human tissues indicated were harvested under identical conditions, and functional hematopoietic progenitor capacity was evaluated at day 0 (de novo–isolated tissues) and again at day 5 of culture in HGF conditions with or without BMP-4 or BMP-4 together with EPO. (A) Developmental potential of hematopoietic progenitors was compared among various human tissues by enumerating colony types as a measure of hematopoietic lineage commitment that include erythroid (BFU-E), monocytic (CFU-M), granulocytic (CFU-G), and myelocytic (CFU-GM). Composition of CFU types generated for each tissue is expressed as mean percentage ± SEM (n = 5) of the total colonies for muscle (gray), neural (white), and blood (black) cells cultured in essential media containing HGF (i), BMP-4 (ii), or BMP-4 and EPO in combination (iii). Single and double asterisks indicate substantial differences within measured groups ofP < .01, (n = 5). (B) Fold changes in total hematopoietic progenitor expansion was compared in response to culture conditions indicated and compared with day 0 of cultured fetal blood (i), muscle (ii), and neural (iii) cells. Each graph shows the increase in progenitors after culture in essential media containing HGF (●), BMP-4 (♦), or BMP-4 and EPO (▪). (C) Fold expansion of fetal blood cultured for 5 days in the absence or presence of cells derived from fetal muscle (i) or fetal neural (ii) tissue using cytokine combinations indicated. Cells were cocultured using 5.0 × 105 fetal blood cells to equal numbers of fetal muscle or neural cells to maintain the same cell density used in previous experiments shown in panel B. Insets show the relative fold expansion to fetal blood cells cultured alone compared with the various coculture treatments. (D) Hematopoietic progenitors assayed from single cells suspensions of muscle (i), neural (ii), and blood (iii) in the absence or presence of BMP-4 directly added to methylcellulose used in this clonal assay. Graphs show number of clonogenic progenitor of de novo–isolated tissues in the absence of BMP-4 (control) shown standardized as 100% and in the presence of BMP-4 as a percentage relative to control ± SEM. Results are based on a total of 5 independent samples. Karen E. Jay et al. Blood 2002;100:3193-3202 ©2002 by American Society of Hematology

Isolation of AC133+ and AC133−subsets from human embryonic tissues Isolation of AC133+ and AC133−subsets from human embryonic tissues.Human fetal neural cells were stained with human-specific antibodies raised to the prominin AC133, CD34, and CD45. Isolation of AC133+ and AC133−subsets from human embryonic tissues.Human fetal neural cells were stained with human-specific antibodies raised to the prominin AC133, CD34, and CD45. AC133+ and AC133− cells were isolated according to sorting gates shown in Figure 1, from the population of cells devoid of CD34 and the hematopoietic marker CD45. (A) Purified subpopulations of AC133+CD34−CD45− (Bi) and AC133−CD45−CD34− (Bii) cells from human neural tissue were cultured under serum-free conditions shown to induce neural hematopoiesis. Magnification × 200 (Bi) and × 400 (Bii). Hematopoietic colonies of multiple lineages were detected from AC133+CD34−CD45− cells. The composition of colonies was similar to that shown in Figure 4. (C) Quantitative analysis of hematopoietic colonies arising from either AC133+ or AC133− subsets from the CD34−CD45− population. Cells were cultured in HGF with BMP-4 and EPO and were then collected and plated into colony-forming assays. Colonies were scored after 12 to 14 days and shown as the average number of colonies per 50 000 cell input ± SEM. Averages shown are based on 4 independent samples. Karen E. Jay et al. Blood 2002;100:3193-3202 ©2002 by American Society of Hematology