Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 1653, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395882

ABSTRACT

Hematopoietic stem cells (HSCs) produce all essential cellular components of the blood. Stromal cell lines supporting HSCs follow a vascular smooth muscle cell (vSMC) differentiation pathway, suggesting that some hematopoiesis-supporting cells originate from vSMC precursors. These pericyte-like precursors were recently identified in the aorta-gonad-mesonephros (AGM) region; however, their role in the hematopoietic development in vivo remains unknown. Here, we identify a subpopulation of NG2+Runx1+ perivascular cells that display a sclerotome-derived vSMC transcriptomic profile. We show that deleting Runx1 in NG2+ cells impairs the hematopoietic development in vivo and causes transcriptional changes in pericytes/vSMCs, endothelial cells and hematopoietic cells in the murine AGM. Importantly, this deletion leads also to a significant reduction of HSC reconstitution potential in the bone marrow in vivo. This defect is developmental, as NG2+Runx1+ cells were not detected in the adult bone marrow, demonstrating the existence of a specialised pericyte population in the HSC-generating niche, unique to the embryo.


Subject(s)
Endothelial Cells , Muscle, Smooth, Vascular , Mice , Animals , Muscle, Smooth, Vascular/metabolism , Endothelial Cells/metabolism , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Hematopoiesis/genetics , Mesonephros , Gonads/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism
2.
STAR Protoc ; 4(1): 102016, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36640365

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) can differentiate into osteoblasts under appropriate conditions. PDGFRß signaling controls MSC osteogenic potential both transcriptomically and in culture. Here, we present a "computer to the bench" protocol to analyze changes in MSC osteogenic potential at transcriptomic and cellular level in the absence of PDGFRß. We detail the preparation of cells from mouse embryos, the analysis of transcriptomic changes from single-cell RNA-sequencing data, the procedure for MSC derivation and culture, and an osteogenic assay for functional validation. For complete details on the use and execution of this protocol, please refer to Sá da Bandeira et al. (2022).1.


Subject(s)
Mesenchymal Stem Cells , Transcriptome , Animals , Mice , Transcriptome/genetics , Cell Differentiation/genetics , Osteogenesis/genetics , Gene Expression Profiling
3.
Cell Rep ; 40(3): 111114, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858557

ABSTRACT

Hematopoietic stem cell (HSC) generation in the aorta-gonad-mesonephros region requires HSC specification signals from the surrounding microenvironment. In zebrafish, PDGF-B/PDGFRß signaling controls hematopoietic stem/progenitor cell (HSPC) generation and is required in the HSC specification niche. Little is known about murine HSPC specification in vivo and whether PDGF-B/PDGFRß is involved. Here, we show that PDGFRß is expressed in distinct perivascular stromal cell layers surrounding the mid-gestation dorsal aorta, and its deletion impairs hematopoiesis. We demonstrate that PDGFRß+ cells play a dual role in murine hematopoiesis. They act in the aortic niche to support HSPCs, and in addition, PDGFRß+ embryonic precursors give rise to a subset of HSPCs that persist into adulthood. These findings provide crucial information for the controlled production of HSPCs in vitro.


Subject(s)
Mesonephros , Zebrafish , Animals , Hematopoiesis , Hematopoietic Stem Cells , Mice , Receptor, Platelet-Derived Growth Factor beta , Stromal Cells
4.
Stem Cells Transl Med ; 11(1): 35-43, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35641167

ABSTRACT

The vascular wall is comprised of distinct layers controlling angiogenesis, blood flow, vessel anchorage within organs, and cell and molecule transit between blood and tissues. Moreover, some blood vessels are home to essential stem-like cells, a classic example being the existence in the embryo of hemogenic endothelial cells at the origin of definitive hematopoiesis. In recent years, microvascular pericytes and adventitial perivascular cells were observed to include multi-lineage progenitor cells involved not only in organ turnover and regeneration but also in pathologic remodeling, including fibrosis and atherosclerosis. These perivascular mesodermal elements were identified as native forerunners of mesenchymal stem cells. We have presented in this brief review our current knowledge on vessel wall-associated tissue remodeling cells with respect to discriminating phenotypes, functional diversity in health and disease, and potential therapeutic interest.


Subject(s)
Mesenchymal Stem Cells , Peripheral Blood Stem Cells , Endothelial Cells , Humans , Mesenchymal Stem Cells/physiology , Pericytes , Stem Cells/physiology
5.
Nat Metab ; 4(1): 123-140, 2022 01.
Article in English | MEDLINE | ID: mdl-35102339

ABSTRACT

Vascular mural cells (vMCs) play an essential role in the development and maturation of the vasculature by promoting vessel stabilization through their interactions with endothelial cells. Whether endothelial metabolism influences mural cell recruitment and differentiation is unknown. Here, we show that the oxidative pentose phosphate pathway (oxPPP) in endothelial cells is required for establishing vMC coverage of the dorsal aorta during early vertebrate development in zebrafish and mice. We demonstrate that laminar shear stress and blood flow maintain oxPPP activity, which in turn, promotes elastin expression in blood vessels through production of ribose-5-phosphate. Elastin is both necessary and sufficient to drive vMC recruitment and maintenance when the oxPPP is active. In summary, our work demonstrates that endothelial cell metabolism regulates blood vessel maturation by controlling vascular matrix composition and vMC recruitment.


Subject(s)
Blood Vessels/cytology , Blood Vessels/metabolism , Extracellular Matrix/metabolism , Oxidative Phosphorylation , Pentose Phosphate Pathway , Animals , Biomarkers , Elastin/biosynthesis , Elastin/genetics , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gene Expression , Genes, Reporter , Glucose/metabolism , Hemodynamics , Mice , Mice, Knockout , Models, Biological , Oxidative Stress , Pentosephosphates/metabolism , Zebrafish
6.
Methods Mol Biol ; 2235: 27-35, 2021.
Article in English | MEDLINE | ID: mdl-33576968

ABSTRACT

Pericytes are mural cells closely associated with endothelial cells in capillaries and microvessels. They are precursors of mesenchymal stem/stromal cells that have historically been retrospectively characterized in culture. We established a protocol, described in this chapter, to characterize and isolate pericytes from multiple human organs by flow cytometry and fluorescence-activated cell sorting. This prospective purification of pericytes brings us a step forward in the development of strategies for their use in the clinic.


Subject(s)
Flow Cytometry/methods , Pericytes/cytology , Pericytes/transplantation , Capillaries/cytology , Cell Culture Techniques/methods , Cell Separation/methods , Cells, Cultured , Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Microvessels/cytology , Pericytes/metabolism , Phenotype
7.
Methods Mol Biol ; 2235: 37-45, 2021.
Article in English | MEDLINE | ID: mdl-33576969

ABSTRACT

Pericytes are found in all vascularized organs and are defined anatomically as perivascular cells that closely surround endothelial cells in capillaries and microvessels and are embedded within the same basement membrane. They have been shown to have diverse physiological and pathological functions including regulation of blood pressure, and tissue regeneration and scarring. Fundamental to understanding the role these cells play in these diverse processes is the ability to accurately identify and localize them in vivo. To do this, we have developed multicolor immunohistochemistry protocols described in this chapter.


Subject(s)
Immunohistochemistry/methods , Pericytes/cytology , Pericytes/transplantation , Capillaries/cytology , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Humans , Microvessels/cytology , Pericytes/metabolism , Phenotype
8.
Article in English | MEDLINE | ID: mdl-32185170

ABSTRACT

Mesenchymal stem cells are culture-derived mesodermal progenitors isolatable from all vascularized tissues. In spite of multiple fundamental, pre-clinical and clinical studies, the native identity and role in tissue repair of MSCs have long remained elusive, with MSC selection in vitro from total cell suspensions essentially unchanged as a mere primary culture for half a century. Recent investigations have helped understand the tissue origin of these progenitor cells, and uncover alternative effects of MSCs on tissue healing via growth factor secretion and interaction with the immune system. In this review, we describe current trends in MSC biology and discuss how these may improve the use of these therapeutic cells in tissue engineering and regenerative medicine.

10.
PLoS One ; 12(5): e0177308, 2017.
Article in English | MEDLINE | ID: mdl-28489940

ABSTRACT

For over 15 years, human subcutaneous adipose tissue has been recognized as a rich source of tissue resident mesenchymal stem/stromal cells (MSC). The isolation of perivascular progenitor cells from human adipose tissue by a cell sorting strategy was first published in 2008. Since this time, the interest in using pericytes and related perivascular stem/stromal cell (PSC) populations for tissue engineering has significantly increased. Here, we describe a set of experiments identifying, isolating and characterizing PSC from canine tissue (N = 12 canine adipose tissue samples). Results showed that the same antibodies used for human PSC identification and isolation are cross-reactive with canine tissue (CD45, CD146, CD34). Like their human correlate, canine PSC demonstrate characteristics of MSC including cell surface marker expression, colony forming unit-fibroblast (CFU-F) inclusion, and osteogenic differentiation potential. As well, canine PSC respond to osteoinductive signals in a similar fashion as do human PSC, such as the secreted differentiation factor NEL-Like Molecule-1 (NELL-1). Nevertheless, important differences exist between human and canine PSC, including differences in baseline osteogenic potential. In summary, canine PSC represent a multipotent mesenchymogenic cell source for future translational efforts in tissue engineering.


Subject(s)
Adipose Tissue/cytology , Cell Separation , Osteogenesis , Stromal Cells/cytology , Tissue Engineering , Animals , Bone and Bones/cytology , Bone and Bones/physiology , Calcium-Binding Proteins , Cell Differentiation , Cell Separation/methods , Cells, Cultured , Dogs , Fibroblast Growth Factor 2/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Platelet-Derived Growth Factor/metabolism , Recombinant Proteins/metabolism , Stromal Cells/metabolism , Tissue Engineering/methods , Vascular Endothelial Growth Factor A/metabolism
11.
Development ; 143(24): 4571-4581, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27965438

ABSTRACT

Not all hematopoietic stem cells (HSCs) are alike. They differ in their physical characteristics such as cell cycle status and cell surface marker phenotype, they respond to different extrinsic signals, and they have different lineage outputs following transplantation. The growing body of evidence that supports heterogeneity within HSCs, which constitute the most robust cell fraction at the foundation of the adult hematopoietic system, is currently of great interest and raises questions as to why HSC subtypes exist, how they are generated and whether HSC heterogeneity affects leukemogenesis or treatment options. This Review provides a developmental overview of HSC subtypes during embryonic, fetal and adult stages of hematopoiesis and discusses the possible origins and consequences of HSC heterogeneity.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/classification , Hematopoietic Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Lineage/physiology , Endothelial Cells/cytology , Humans , Stem Cell Niche
12.
Stem Cell Reports ; 6(3): 383-95, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26923823

ABSTRACT

Hematopoietic stem cells (HSC), the self-renewing cells of the adult blood differentiation hierarchy, are generated during embryonic stages. The first HSCs are produced in the aorta-gonad-mesonephros (AGM) region of the embryo through endothelial to a hematopoietic transition. BMP4 and Hedgehog affect their production and expansion, but it is unknown whether they act to affect the same HSCs. In this study using the BRE GFP reporter mouse strain that identifies BMP/Smad-activated cells, we find that the AGM harbors two types of adult-repopulating HSCs upon explant culture: One type is BMP-activated and the other is a non-BMP-activated HSC type that is indirectly controlled by Hedgehog signaling through the VEGF pathway. Transcriptomic analyses demonstrate that the two HSC types express distinct but overlapping genetic programs. These results revealing the bifurcation in HSC types at early embryonic stages in the AGM explant model suggest that their development is dependent upon the signaling molecules in the microenvironment.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Hedgehog Proteins/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Smad Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism
13.
Circ Res ; 118(5): 822-33, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26838788

ABSTRACT

RATIONALE: Pulmonary arterial hypertension is characterized by vascular remodeling and neomuscularization. PW1(+) progenitor cells can differentiate into smooth muscle cells (SMCs) in vitro. OBJECTIVE: To determine the role of pulmonary PW1(+) progenitor cells in vascular remodeling characteristic of pulmonary arterial hypertension. METHODS AND RESULTS: We investigated their contribution during chronic hypoxia-induced vascular remodeling in Pw1(nLacZ+/-) mouse expressing ß-galactosidase in PW1(+) cells and in differentiated cells derived from PW1(+) cells. PW1(+) progenitor cells are present in the perivascular zone in rodent and human control lungs. Using progenitor markers, 3 distinct myogenic PW1(+) cell populations were isolated from the mouse lung of which 2 were significantly increased after 4 days of chronic hypoxia. The number of proliferating pulmonary PW1(+) cells and the proportion of ß-gal(+) vascular SMC were increased, indicating a recruitment of PW1(+) cells and their differentiation into vascular SMC during early chronic hypoxia-induced neomuscularization. CXCR4 inhibition using AMD3100 prevented PW1(+) cells differentiation into SMC but did not inhibit their proliferation. Bone marrow transplantation experiments showed that the newly formed ß-gal(+) SMC were not derived from circulating bone marrow-derived PW1(+) progenitor cells, confirming a resident origin of the recruited PW1(+) cells. The number of pulmonary PW1(+) cells was also increased in rats after monocrotaline injection. In lung from pulmonary arterial hypertension patients, PW1-expressing cells were observed in large numbers in remodeled vascular structures. CONCLUSIONS: These results demonstrate the existence of a novel population of resident SMC progenitor cells expressing PW1 and participating in pulmonary hypertension-associated vascular remodeling.


Subject(s)
Hypertension, Pulmonary/metabolism , Kruppel-Like Transcription Factors/biosynthesis , Muscle, Smooth, Vascular/metabolism , Stem Cells/metabolism , Vascular Remodeling/physiology , Animals , Cells, Cultured , Humans , Hypertension, Pulmonary/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Rats , Stem Cells/pathology
15.
Nat Commun ; 6: 8040, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26282601

ABSTRACT

Adult haematopoiesis is the outcome of distinct haematopoietic stem cell (HSC) subtypes with self-renewable repopulating ability, but with different haematopoietic cell lineage outputs. The molecular basis for this heterogeneity is largely unknown. BMP signalling regulates HSCs as they are first generated in the aorta-gonad-mesonephros region, but at later developmental stages, its role in HSCs is controversial. Here we show that HSCs in murine fetal liver and the bone marrow are of two types that can be prospectively isolated--BMP activated and non-BMP activated. Clonal transplantation demonstrates that they have distinct haematopoietic lineage outputs. Moreover, the two HSC types differ in intrinsic genetic programs, thus supporting a role for the BMP signalling axis in the regulation of HSC heterogeneity and lineage output. Our findings provide insight into the molecular control mechanisms that define HSC types and have important implications for reprogramming cells to HSC fate and treatments targeting distinct HSC types.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/physiology , Signal Transduction/physiology , Animals , Benzofurans , Bone Morphogenetic Proteins/genetics , Embryo, Mammalian , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Quinolines
16.
J Vis Exp ; (90): e51195, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25177794

ABSTRACT

Since the discovery of mesenchymal stem/stromal cells (MSCs), the native identity and localization of MSCs have been obscured by their retrospective isolation in culture. Recently, using fluorescence-activated cell sorting (FACS), we and other researchers prospectively identified and purified three subpopulations of multipotent precursor cells associated with the vasculature of human skeletal muscle. These three cell populations: myogenic endothelial cells (MECs), pericytes (PCs), and adventitial cells (ACs), are localized respectively to the three structural layers of blood vessels: intima, media, and adventitia. All of these human blood-vessel-derived stem cell (hBVSC) populations not only express classic MSC markers but also possess mesodermal developmental potentials similar to typical MSCs. Previously, MECs, PCs, and ACs have been isolated through distinct protocols and subsequently characterized in separate studies. The current isolation protocol, through modifications to the isolation process and adjustments in the selective cell surface markers, allows us to simultaneously purify all three hBVSC subpopulations by FACS from a single human muscle biopsy. This new method will not only streamline the isolation of multiple BVSC subpopulations but also facilitate future clinical applications of hBVSCs for distinct therapeutic purposes.


Subject(s)
Adventitia/cytology , Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Muscle, Skeletal/blood supply , Pericytes/cytology , Tunica Intima/cytology , Humans
17.
Stem Cell Res ; 12(1): 24-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24141110

ABSTRACT

Hypoxia affects many physiologic processes during early stages of mammalian ontogeny, particularly placental and vascular development. In the adult, the hypoxic bone marrow microenvironment plays a role in regulating hematopoietic stem cell (HSC) function. HSCs are generated from the major vasculature of the embryo, but whether the hypoxic response affects the generation of these HSCs is as yet unknown. Here we examined whether Hypoxia Inducible Factor1-alpha (HIF1α), a key modulator of the response to hypoxia, is essential for HSC development. We found hypoxic cells in embryonic tissues that generate and expand hematopoietic cells (aorta, placenta and fetal liver), and specifically aortic endothelial and hematopoietic cluster cells. A Cre/loxP conditional knockout (cKO) approach was taken to delete HIF1α in Vascular Endothelial-Cadherin expressing endothelial cells, the precursors to definitive hematopoietic cells. Functional assays show that HSC and hematopoietic progenitor cells (HPCs) are significantly reduced in cKO aorta and placenta. Moreover, decreases in phenotypic aortic hematopoietic cluster cells in cKO embryos indicate that HIF1α is necessary for generation and/or expansion of HPCs and HSCs. cKO adult BM HSCs are also affected under transplantation conditions. Thus, HIF1α is a regulator of HSC generation and function beginning at the earliest embryonic stages.


Subject(s)
Cell Hypoxia/physiology , Hematopoietic Stem Cells/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Aorta/cytology , Cadherins/metabolism , Cell Separation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Fetus/cytology , Hematopoietic Stem Cell Transplantation , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver/cytology , Mice , Mice, Inbred C57BL , Placenta/cytology , Pregnancy , Transplantation, Homologous
18.
Stem Cell Res Ther ; 4(4): 95, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23953698

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) are heterogeneous. A fraction of these cells constitute multipotent cells that can self-renew and mainly give rise to mesodermal lineage cells such as adipocytes, osteocytes and chondrocytes. The ability of MSCs to differentiate into endothelial cells remains controversial. Isolation and in vitro manipulation of MSCs before clinical application are important steps. High numbers of MSCs are needed, requiring the in vitro expansion of these clinically important cells. To this end, a well-controlled procedure for MSC isolation and maintenance in culture is necessary.


Subject(s)
Cell Culture Techniques/methods , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/cytology , Humans
19.
Cytometry A ; 83(8): 714-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23818229

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) are adult multipotent progenitors of great promise for cell therapy. MSCs can mediate tissue regeneration, immunomodulation, and hematopoiesis support. Despite the unique properties of MSCs and their broad range of potential clinical applications, the very nature of these cells has been uncertain. Furthermore, MSCs are heterogeneous and only defined subpopulations of these are endowed with the particular abilities to sustain hematopoietic stem cells, regulate immune responses, or differentiate into mesodermal cell lineages. It is becoming evident that current criteria used to define cultured polyclonal MSCs (expression of nonspecific markers and in vitro mesodermal differentiation) are not sufficient to fully understand and exploit the potential of these cells. Here, we describe how flow cytometry has been used to reveal a perivascular origin of MSCs. As a result, the prospective purification of MSCs and specialized subsets thereof is now possible, and the clinical use of purified autologous MSCs is now within reach.


Subject(s)
Flow Cytometry , Mesenchymal Stem Cells/metabolism , Adipose Tissue, White/cytology , Animals , Antigens, CD/metabolism , Blood Vessels/cytology , Cell Separation , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Regenerative Medicine
20.
Stem Cells ; 31(2): 305-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23165704

ABSTRACT

Human microvascular pericytes (CD146(+)/34(-)/45(-)/56(-)) contain multipotent precursors and repair/regenerate defective tissues, notably skeletal muscle. However, their ability to repair the ischemic heart remains unknown. We investigated the therapeutic potential of human pericytes, purified from skeletal muscle, for treating ischemic heart disease and mediating associated repair mechanisms in mice. Echocardiography revealed that pericyte transplantation attenuated left ventricular dilatation and significantly improved cardiac contractility, superior to CD56+ myogenic progenitor transplantation, in acutely infarcted mouse hearts. Pericyte treatment substantially reduced myocardial fibrosis and significantly diminished infiltration of host inflammatory cells at the infarct site. Hypoxic pericyte-conditioned medium suppressed murine fibroblast proliferation and inhibited macrophage proliferation in vitro. High expression by pericytes of immunoregulatory molecules, including interleukin-6, leukemia inhibitory factor, cyclooxygenase-2, and heme oxygenase-1, was sustained under hypoxia, except for monocyte chemotactic protein-1. Host angiogenesis was significantly increased. Pericytes supported microvascular structures in vivo and formed capillary-like networks with/without endothelial cells in three-dimensional cocultures. Under hypoxia, pericytes dramatically increased expression of vascular endothelial growth factor-A, platelet-derived growth factor-ß, transforming growth factor-ß1 and corresponding receptors while expression of basic fibroblast growth factor, hepatocyte growth factor, epidermal growth factor, and angiopoietin-1 was repressed. The capacity of pericytes to differentiate into and/or fuse with cardiac cells was revealed by green fluorescence protein labeling, although to a minor extent. In conclusion, intramyocardial transplantation of purified human pericytes promotes functional and structural recovery, attributable to multiple mechanisms involving paracrine effects and cellular interactions.


Subject(s)
Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/pathology , Pericytes/transplantation , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Culture Techniques , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Fibrosis/prevention & control , Gene Expression , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardium/metabolism , Neovascularization, Physiologic , Pericytes/physiology , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , Regeneration/physiology , Transplantation, Heterologous , Ultrasonography , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...