Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
J Biol Chem ; 285(46): 35645-53, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20826809

ABSTRACT

Transplantation of mesenchymal stem cells (MSCs) is a promising therapy for ischemic injury; however, inadequate survival of implanted cells in host tissue is a substantial impediment in the progress of cellular therapy. Secreted Frizzled-related protein 2 (sFRP2) has recently been highlighted as a key mediator of MSC-driven myocardial and wound repair. Notably, sFRP2 mediates significant enhancement of MSC engraftment in vivo. We hypothesized that sFRP2 improves MSC engraftment by modulating self-renewal through increasing stem cell survival and by inhibiting differentiation. In previous studies we demonstrated that sFRP2-expressing MSCs exhibited an increased proliferation rate. In the current study, we show that sFRP2 also decreased MSC apoptosis and inhibited both osteogenic and chondrogenic lineage commitment. sFRP2 activity occurred through the inhibition of both Wnt and bone morphogenic protein (BMP) signaling pathways. sFRP2-mediated inhibition of BMP signaling, as assessed by levels of pSMAD 1/5/8, was independent of its effects on the Wnt pathway. We further hypothesized that sFRP2 inhibition of MSC lineage commitment may reduce heterotopic osteogenic differentiation within the injured myocardium, a reported adverse side effect. Indeed, we found that sFRP2-MSC-treated hearts and wound tissue had less ectopic calcification. This work provides important new insight into the mechanisms by which sFRP2 increases MSC self-renewal leading to superior tissue engraftment and enhanced wound healing.


Subject(s)
Membrane Proteins/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/surgery , Signal Transduction , Animals , Apoptosis , Bone Morphogenetic Proteins/metabolism , Calcinosis/etiology , Calcinosis/surgery , Cell Differentiation , Cell Proliferation , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Glucuronidase/deficiency , Glucuronidase/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Myocardial Infarction/complications , Myocardial Infarction/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Wnt Proteins/metabolism
2.
Cancer Res ; 69(1): 338-48, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19118019

ABSTRACT

Microenvironmental cues instruct infiltrating tumor-associated myeloid cells to drive malignant progression. A subpopulation of tumor-associated myeloid cells coexpressing endothelial and myeloid markers, although rare in peripheral blood, are primarily associated with tumors where they enhance tumor growth and angiogenesis. These biphenotypic vascular leukocytes result from the endothelial differentiation of myeloid progenitors, a process regulated by tumor necrosis factor (TNF)alpha in vitro. An in vivo increase in tumor-derived TNFalpha expression promoted tumor growth and vascularity of mouse melanoma, lung cancer, and mammary tumors. Notably, tumor growth was accompanied by a significant increase in myeloid/endothelial biphenotypic populations. TNFalpha-associated tumor growth, vascularity, and generation of tumor vascular leukocytes in mouse melanoma tumors were dependent on intact host TNFalpha receptors. Importantly, TNFalpha-expressing tumors did not exhibit increased inflammation over control tumors, suggesting a unique action related to myeloid to endothelial differentiation. Our studies suggest that TNFalpha constitutes a tumor microenvironment signal that biases recruited monocytes toward a proangiogenic/provasculogenic myeloid/endothelial phenotype.


Subject(s)
Carcinoma, Lewis Lung/pathology , Endothelial Cells/pathology , Monocytes/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Cell Differentiation/drug effects , Cell Growth Processes/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Lipopolysaccharide Receptors/biosynthesis , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Monocytes/drug effects , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
3.
Proc Natl Acad Sci U S A ; 105(47): 18366-71, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19017790

ABSTRACT

Cell-based therapies, using multipotent mesenchymal stem cells (MSCs) for organ regeneration, are being pursued for cardiac disease, orthopedic injuries and biomaterial fabrication. The molecular pathways that regulate MSC-mediated regeneration or enhance their therapeutic efficacy are, however, poorly understood. We compared MSCs isolated from MRL/MpJ mice, known to demonstrate enhanced regenerative capacity, to those from C57BL/6 (WT) mice. Compared with WT-MSCs, MRL-MSCs demonstrated increased proliferation, in vivo engraftment, experimental granulation tissue reconstitution, and tissue vascularity in a murine model of repair stimulation. The MRL-MSCs also reduced infarct size and improved function in a murine myocardial infarct model compared with WT-MSCs. Genomic and functional analysis indicated a downregulation of the canonical Wnt pathway in MRL-MSCs characterized by significant up-regulation of specific secreted frizzled-related proteins (sFRPs). Specific knockdown of sFRP2 by shRNA in MRL-MSCs decreased their proliferation and their engraftment in and the vascular density of MRL-MSC-generated experimental granulation tissue. These results led us to generate WT-MSCs overexpressing sFRP2 (sFRP2-MSCs) by retroviral transduction. sFRP2-MSCs maintained their ability for multilineage differentiation in vitro and, when implanted in vivo, recapitulated the MRL phenotype. Peri-infarct intramyocardial injection of sFRP2-MSCs resulted in enhanced engraftment, vascular density, reduced infarct size, and increased cardiac function after myocardial injury in mice. These findings implicate sFRP2 as a key molecule for the biogenesis of a superior regenerative phenotype in MSCs.


Subject(s)
Heart/physiopathology , Membrane Proteins/physiology , Mesenchymal Stem Cells/cytology , Regeneration/physiology , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Wound Healing
4.
J Hum Lact ; 24(1): 58-68, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18281357

ABSTRACT

The Institute of Medicine has issued a call for the implementation of computer-based patient records. The purpose of this overview is to describe the content developed for an electronic health record in an outpatient breastfeeding medicine clinic at a pediatric health care facility. Additional features of the computer system, including electronic prescriptions, printed patient handouts, and follow-up telephone notes, utilized in this setting are also described. As more hospital systems and outpatient offices adopt electronic medical record systems, the authors recommend that lactation personnel work with administrators and computer professionals at their institution to incorporate the special needs of an outpatient lactation clinic into their system.


Subject(s)
Breast Feeding/statistics & numerical data , Medical Records Systems, Computerized , Quality of Health Care , Humans , Outpatients
SELECTION OF CITATIONS
SEARCH DETAIL