Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biomaterials ; 182: 44-57, 2018 11.
Article in English | MEDLINE | ID: mdl-30103171

ABSTRACT

The use of degradable materials is required to address current performance and functionality shortcomings from biologically-derived tissues and non-resorbable synthetic materials used for hernia mesh repair applications. Herein a series of degradable l-valine-co-l-phenylalanine poly(ester urea) (PEU) copolymers were investigated for soft-tissue repair. Poly[(1-VAL-8)0.7-co-(1-PHE-6)0.3] showed the highest uniaxial mechanical properties (332.5 ±â€¯3.5 MPa). Additionally, l-valine-co-l-phenylalanine poly(ester urea)s were blade coated on small intestine submucosa extracellular matrix (SIS-ECM) and found to enhance the burst test mechanical properties of SIS-ECM in composite films (force at break between 102.6 ±â€¯6.5-151.4 ±â€¯11.3 N). Free standing films of l-valine-co-l-phenylalanine PEUs were found to have superior extension at break when compared to SIS-ECM (averages between 1.2 and 1.9 cm and 1.2 cm respectively). Fibroblast (L-929) spreading, proliferation, and improved attachment over control were observed without toxicity in vitro, while a reduced inflammatory response at both 7 and 14 days post-implant was observed for poly[(1-VAL-8)⁠0.7-co-(1-PHE-6)⁠0.3] when compared to polypropylene in an in vivo rat hernia model. These results support the use of PEU copolymers as free-standing films or as composite materials in soft-tissue applications for hernia-repair.


Subject(s)
Absorbable Implants , Biocompatible Materials/chemistry , Herniorrhaphy/methods , Phenylalanine/analogs & derivatives , Polyesters/chemistry , Urea/analogs & derivatives , Valine/analogs & derivatives , Animals , Cell Line , Cell Survival , Elastic Modulus , Hernia/therapy , Materials Testing , Mice , Rats , Urea/chemistry
2.
Proc Natl Acad Sci U S A ; 115(13): E3036-E3044, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29531045

ABSTRACT

Heart failure (HF) following myocardial infarction (MI) is associated with high incidence of cardiac arrhythmias. Development of therapeutic strategy requires detailed understanding of electrophysiological remodeling. However, changes of ionic currents in ischemic HF remain incompletely understood, especially in translational large-animal models. Here, we systematically measure the major ionic currents in ventricular myocytes from the infarct border and remote zones in a porcine model of post-MI HF. We recorded eight ionic currents during the cell's action potential (AP) under physiologically relevant conditions using selfAP-clamp sequential dissection. Compared with healthy controls, HF-remote zone myocytes exhibited increased late Na+ current, Ca2+-activated K+ current, Ca2+-activated Cl- current, decreased rapid delayed rectifier K+ current, and altered Na+/Ca2+ exchange current profile. In HF-border zone myocytes, the above changes also occurred but with additional decrease of L-type Ca2+ current, decrease of inward rectifier K+ current, and Ca2+ release-dependent delayed after-depolarizations. Our data reveal that the changes in any individual current are relatively small, but the integrated impacts shift the balance between the inward and outward currents to shorten AP in the border zone but prolong AP in the remote zone. This differential remodeling in post-MI HF increases the inhomogeneity of AP repolarization, which may enhance the arrhythmogenic substrate. Our comprehensive findings provide a mechanistic framework for understanding why single-channel blockers may fail to suppress arrhythmias, and highlight the need to consider the rich tableau and integration of many ionic currents in designing therapeutic strategies for treating arrhythmias in HF.


Subject(s)
Action Potentials/physiology , Arrhythmias, Cardiac/physiopathology , Calcium/metabolism , Electrophysiological Phenomena , Heart Failure/physiopathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Animals , Cells, Cultured , Myocytes, Cardiac/cytology , Swine
3.
ACS Biomater Sci Eng ; 4(4): 1346-1356, 2018 Apr 09.
Article in English | MEDLINE | ID: mdl-33418665

ABSTRACT

New polymers are needed to address the shortcomings of commercially available materials for soft tissue repair. Herein, we investigated a series of l-valine-based poly(ester urea)s (PEUs) that vary in monomer composition and the extent of branching as candidate materials for soft tissue repair. The preimplantation Young's moduli (105 ± 30 to 269 ± 12 MPa) for all the PEUs are comparable to those of polypropylene (165 ± 5 MPa) materials currently employed in hernia-mesh repair. The 2% branched poly(1-VAL-8) maintained the highest Young's modulus following 3 months of in vivo implantation (78 ± 34 MPa) when compared to other PEU analogues (20 ± 6-45 ± 5 MPa). Neither the linear or branched PEUs elicited a significant inflammatory response in vivo as noted by less fibrous capsule formation after 3 months of implantation (80 ± 38 to 103 ± 33 µm) relative to polypropylene controls (126 ± 34 µm). Mechanical degradation in vivo over three months, coupled with limited inflammatory response, suggests that l-valine-based PEUs are translationally relevant materials for soft tissue applications.

4.
PLoS One ; 11(4): e0153412, 2016.
Article in English | MEDLINE | ID: mdl-27070546

ABSTRACT

In this study, we investigate the translational potential of a novel combined construct using an FDA-approved decellularized porcine small intestinal submucosa extracellular matrix (SIS-ECM) seeded with human or porcine mesenchymal stem cells (MSCs) for cardiovascular indications. With the emerging success of individual component in various clinical applications, the combination of SIS-ECM with MSCs could provide additional therapeutic potential compared to individual components alone for cardiovascular repair. We tested the in vitro effects of MSC-seeding on SIS-ECM on resultant construct structure/function properties and MSC phenotypes. Additionally, we evaluated the ability of porcine MSCs to modulate recipient graft-specific response towards SIS-ECM in a porcine cardiac patch in vivo model. Specifically, we determined: 1) in vitro loading-capacity of human MSCs on SIS-ECM, 2) effect of cell seeding on SIS-ECM structure, compositions and mechanical properties, 3) effect of SIS-ECM seeding on human MSC phenotypes and differentiation potential, and 4) optimal orientation and dose of porcine MSCs seeded SIS-ECM for an in vivo cardiac application. In this study, histological structure, biochemical compositions and mechanical properties of the FDA-approved SIS-ECM biomaterial were retained following MSCs repopulation in vitro. Similarly, the cellular phenotypes and differentiation potential of MSCs were preserved following seeding on SIS-ECM. In a porcine in vivo patch study, the presence of porcine MSCs on SIS-ECM significantly reduced adaptive T cell response regardless of cell dose and orientation compared to SIS-ECM alone. These findings substantiate the clinical translational potential of combined SIS-ECM seeded with MSCs as a promising therapeutic candidate for cardiac applications.


Subject(s)
Extracellular Matrix/physiology , Heart/physiology , Intestine, Small/cytology , Mesenchymal Stem Cells/cytology , Adaptive Immunity/physiology , Animals , Biocompatible Materials/metabolism , Cell Differentiation/physiology , Cells, Cultured , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Intestine, Small/physiology , Male , Mesenchymal Stem Cells/physiology , Swine , T-Lymphocytes/physiology , Tissue Scaffolds
5.
PLoS One ; 8(12): e82134, 2013.
Article in English | MEDLINE | ID: mdl-24312635

ABSTRACT

Ischemic diseases such as peripheral vascular disease (PVD) affect more than 15% of the general population and in severe cases result in ulcers, necrosis, and limb loss. While the therapeutic delivery of growth factors to promote angiogenesis has been widely investigated, large-scale implementation is limited by strategies to effectively deliver costly recombinant proteins. Multipotent adipose-derived stromal cells (ASC) and progenitor cells from other tissue compartments secrete bioactive concentrations of angiogenic molecules, making cell-based strategies for in situ delivery of angiogenic cytokines an exciting alternative to the use of recombinant proteins. Here, we show that the phospholipid lysophosphatidic acid (LPA) synergistically improves the proangiogenic effects of ASC in ischemia. We found that LPA upregulates angiogenic growth factor production by ASC under two- and three-dimensional in vitro models of serum deprivation and hypoxia (SD/H), and that these factors significantly enhance endothelial cell migration. The concurrent delivery of LPA and ASC in fibrin gels significantly improves vascularization in a murine critical hindlimb ischemia model compared to LPA or ASC alone, thus exhibiting the translational potential of this method. Furthermore, these results are achieved using an inexpensive lipid molecule, which is orders-of-magnitude less costly than recombinant growth factors that are under investigation for similar use. Our results demonstrate a novel strategy for enhancing cell-based strategies for therapeutic angiogenesis, with significant applications for treating ischemic diseases.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Lysophospholipids/pharmacology , Neovascularization, Physiologic/drug effects , Adipose Tissue/cytology , Adult , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation/drug effects , Humans , Ischemia/pathology , Ischemia/physiopathology , Ischemia/therapy , Male , Middle Aged , Receptors, Lysophosphatidic Acid/metabolism , Stem Cells/cytology , Stromal Cells/cytology , Stromal Cells/drug effects , Vascular Endothelial Growth Factor A/genetics
6.
Stem Cells ; 29(11): 1727-37, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21898687

ABSTRACT

A key mechanism for mesenchymal stem cells/bone marrow stromal cells (MSCs) to promote tissue repair is by secretion of soluble growth factors (GFs). Therefore, clinical application could be optimized by a combination of cell and gene therapies, where MSCs are genetically modified to express higher levels of a specific factor. However, it remains unknown how this overexpression may alter the fate of the MSCs. Here, we show effects of overexpressing the growth factors, such as basic fibroblast growth factor (bFGF), platelet derived growth factor B (PDGF-BB), transforming growth factor ß(1) (TGF-ß(1) ), and vascular endothelial growth factor (VEGF), in human bone marrow-derived MSCs. Ectopic expression of bFGF or PDGF-B lead to highly proliferating MSCs and lead to a robust increase in osteogenesis. In contrast, adipogenesis was strongly inhibited in MSCs overexpressing PDGF-B and only mildly affected in MSCs overexpressing bFGF. Overexpression of TGF-ß(1) blocked both osteogenic and adipogenic differentiation while inducing the formation of stress fibers and increasing the expression of the smooth muscle marker calponin-1 and the chondrogenic marker collagen type II. In contrast, MSCs overexpressing VEGF did not vary from control MSCs in any parameters, likely due to the lack of VEGF receptor expression on MSCs. MSCs engineered to overexpress VEGF strongly induced the migration of endothelial cells and enhanced blood flow restoration in a xenograft model of hind limb ischemia. These data support the rationale for genetically modifying MSCs to enhance their therapeutically relevant trophic signals, when safety and efficacy can be demonstrated, and when it can be shown that there are no unwanted effects on their proliferation and differentiation.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell- and Tissue-Based Therapy/methods , Genetic Therapy/methods , Mesenchymal Stem Cells/cytology , Stromal Cells/cytology , Adipogenesis/genetics , Adipogenesis/physiology , Animals , Blotting, Western , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Ischemia/metabolism , Ischemia/therapy , Lentivirus/genetics , Mesenchymal Stem Cells/metabolism , Mice , Mice, Mutant Strains , Osteogenesis/genetics , Osteogenesis/physiology , Real-Time Polymerase Chain Reaction , Stromal Cells/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Ann Thorac Surg ; 90(3): 796-803; discussion 803-4, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20732499

ABSTRACT

BACKGROUND: Mesenchymal stromal cells have been recently isolated from thymus gland tissue discarded after surgical procedures. The role of this novel cell type in heart regeneration has yet to be defined. The purpose of this study was to evaluate the therapeutic potential of human thymus-derived mesenchymal stromal cells using self-organized cardiac tissue as an in vitro platform for quantitative assessment. METHODS: Mesenchymal stromal cells were isolated from discarded thymus tissue from neonates undergoing heart surgery and were incubated in differentiation media to demonstrate multipotency. Neonatal rat cardiomyocytes self-organized into cardiac tissue fibers in a custom culture dish either alone or in combination with varying numbers of mesenchymal stromal cells. A transducer measured force generated by spontaneously contracting self-organized cardiac tissue fibers. Work and power outputs were calculated from force tracings. Immunofluorescence was performed to determine the fate of the thymus-derived mesenchymal stromal cells. RESULTS: Mesenchymal stromal cells were successfully isolated from discarded thymus tissue. After incubation in differentiation media, mesenchymal stromal cells attained the expected phenotypes. Although mesenchymal stromal cells did not differentiate into mature cardiomyocytes, addition of these cells increased the rate of fiber formation, force production, and work and power outputs. Self-organized cardiac tissue containing mesenchymal stromal cells acquired a defined microscopic architecture. CONCLUSIONS: Discarded thymus tissue contains mesenchymal stromal cells, which can augment force production and work and power outputs of self-organized cardiac tissue fibers by several-fold. These findings indicate the potential utility of mesenchymal stromal cells in treating heart failure.


Subject(s)
Heart/anatomy & histology , Mesoderm/cytology , Stromal Cells , Thymus Gland/cytology , Animals , Animals, Newborn , Humans , Rats , Rats, Inbred F344 , Tissue Culture Techniques
8.
J Transl Med ; 8: 24, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20214792

ABSTRACT

UNLABELLED: Human stem cells from adult sources have been shown to contribute to the regeneration of muscle, liver, heart, and vasculature. The mechanisms by which this is accomplished are, however, still not well understood. We tested the engraftment and regenerative potential of human umbilical cord blood-derived ALDH(hi)Lin(-), and ALDH(lo)Lin(-) cells following transplantation to NOD/SCID or NOD/SCID beta2m null mice with experimentally induced acute myocardial infarction. We used combined nanoparticle labeling and whole organ fluorescent imaging to detect human cells in multiple organs 48 hours post transplantation. Engraftment and regenerative effects of cell treatment were assessed four weeks post transplantation. We found that ALDH(hi)Lin(-) stem cells specifically located to the site of injury 48 hours post transplantation and engrafted the infarcted heart at higher frequencies than ALDH(lo)Lin(-) committed progenitor cells four weeks post transplantation. We found no donor derived cardiomyocytes and few endothelial cells of donor origin. Cell treatment was not associated with any detectable functional improvement at the four week endpoint. There was, however, a significant increase in vascular density in the central infarct zone of ALDH(hi)Lin(-) cell-treated mice, as compared to PBS and ALDH(lo)Lin(-) cell-treated mice. CONCLUSIONS: Our data indicate that adult human stem cells do not become a significant part of the regenerating tissue, but rapidly home to and persist only temporarily at the site of hypoxic injury to exert trophic effects on tissue repair thereby enhancing vascular recovery.


Subject(s)
Adult Stem Cells/enzymology , Aldehyde Dehydrogenase/metabolism , Fetal Blood , Myocardial Infarction , Neovascularization, Physiologic/physiology , Animals , Cell Lineage , Cell Separation , Fetal Blood/cytology , Fetal Blood/enzymology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/cytology , Myocardium/metabolism , Regeneration/physiology , Stem Cell Transplantation
9.
Hum Gene Ther ; 20(4): 337-49, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19281432

ABSTRACT

Retroviral vector-mediated gene transfer has been used successfully in clinical gene therapy. Cells of the hematopoietic lineages, however, remain difficult to transduce, although precoating of culture vessels with the fibronectin fragment CH-296 may improve transduction efficiency. Alternatively, low-speed centrifugation of vector-containing supernatant onto culture vessels may improve transduction efficiency in the absence of CH-296 preloading. Using the NIH/3T3-derived Moloney murine leukemia virus-based packaging cell lines PG13, PA317, and PT67, we here show that preloading by low-speed centrifugation improves transduction efficiency in a packaging cell subclone-dependent manner. Preloading by centrifugation, however, cannot generally replace CH-296 and we obtained the overall highest transduction levels when combining centrifugation and CH-296 precoating. We found, moreover, that the factor responsible for high susceptibility to preloading in our PG13-derived vector supernatant was transferable to a PA317-derived vector supernatant with low susceptibility to preloading. Furthermore, our PA317, PG13, and PT67 subclones shed into their supernatants variable amounts of fibronectin. This soluble fibronectin formed aggregates of various sizes and generated complexes with vector particles. The fibronectin-vector complexes readily sedimented onto culture vessels and copurified after fibronectin-specific affinity purification of vector-containing supernatants. Finally, vector supernatant from 293T cells, which barely produce fibronectin, was not susceptible to preloading. The susceptibility to preloading by centrifugation thus appears to be dependent both on the specific packaging cell line and on the association of vector particles and packaging cell-produced fibronectin. Rigorous screening of individual vector-containing supernatants is therefore required to identify optimal transduction conditions for retroviral gene transfer.


Subject(s)
Fibronectins/metabolism , Genetic Vectors/genetics , Retroviridae/genetics , Transduction, Genetic , Virus Assembly , Animals , Cell Line , Centrifugation , Chromatography, Affinity , Clone Cells , Humans , Mice , Time Factors , Tissue Culture Techniques , Virion/isolation & purification
10.
Stem Cells Dev ; 18(6): 845-56, 2009.
Article in English | MEDLINE | ID: mdl-18991484

ABSTRACT

Cell-based regenerative therapy may be useful for treatment of acute myocardial infarction (AMI). Animal xenograft models are ideally suited for preclinical studies evaluating prospective treatment regimes, identifying candidate human cell populations, and gaining mechanistic insight. Here we address whether the athymic nude rat is suitable as a xenograft model for the study of human CD34+ mobilized peripheral blood stem cells (M-PBSCs) in the repair of AMI. We injected human donor cells into the infarct border of athymic nude rats with surgically induced AMI and evaluated engraftment and functional improvement. We found no human engraftment by immunofluorescence staining at 14 days after transplantation or functional improvement at days 2 and 14 compared to controls. The lack of long-term human engraftment was furthermore confirmed in a time series study analyzing animals at 0, 24, 48, 72, and 96 h after transplantation. Although we found fluorescent microbeads coinjected with human CD34+ M-PBSCs at all time points, the number of donor cells rapidly declined and became undetectable at 96 h. CD34+ M-PBSCs from the same donor used to treat athymic nude rat hearts engrafted the bone marrow of nonobese diabetic/severe combined immunodeficient mice 8-10 weeks after transplantation. In conclusion, human CD34+ M-PBSCs with confirmed hematopoietic engraftment potential rapidly disappeared from the site of injury following intramyocardial transplantation in the athymic nude rat AMI model.


Subject(s)
Antigens, CD34/metabolism , Health , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Myocardial Infarction/therapy , Tissue Donors , Animals , Flow Cytometry , Fluorescence , Hematopoiesis , Humans , Kinetics , Leukocyte Common Antigens/metabolism , Mice , Mice, SCID , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Rats , Rats, Nude , Time Factors , Ultrasonography , Ventricular Function, Left
11.
Circulation ; 116(18): 2053-61, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17938286

ABSTRACT

BACKGROUND: Signs of preceding episodes of plaque rupture and smooth muscle cell (SMC)-mediated healing are common in atherosclerotic plaques, but the source of the healing SMCs is unknown. Recent studies suggest that activated platelets adhering to sites of injury recruit neointimal SMCs from circulating bone marrow-derived progenitor cells. Here, we analyzed the contribution of this mechanism to plaque healing after spontaneous and mechanical plaque disruption in apolipoprotein E knockout (apoE-/-) mice. METHODS AND RESULTS: To determine the origin of SMCs after spontaneous plaque disruption, irradiated 18-month-old apoE-/- mice were reconstituted with bone marrow cells from enhanced green fluorescent protein (eGFP) transgenic apoE-/- mice and examined when they died up to 9 months later. Plaque hemorrhage, indicating previous plaque disruption, was widely present, but no bone marrow-derived eGFP+ SMCs were detected. To examine the origin of healing SMCs in a model that recapitulates more features of human plaque rupture and healing, we developed a mechanical technique that produced consistent plaque disruption, superimposed thrombosis, and SMC-mediated plaque healing in apoE-/- mice. Mechanical plaque disruption was produced in irradiated apoE-/- mice reconstituted with eGFP+ apoE-/- bone marrow cells and in carotid bifurcations cross-grafted between apoE-/- and eGFP+ apoE-/- mice. Apart from few non-graft-derived SMCs near the anastomosis site in 1 transplanted carotid bifurcation, no SMCs originating from outside the local arterial segment were detected in healed plaques. CONCLUSIONS: Healing SMCs after atherosclerotic plaque disruption are derived entirely from the local arterial wall and not circulating progenitor cells in apoE-/- mice.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Carotid Arteries/cytology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Smooth Muscle/pathology , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/pathology
12.
Arterioscler Thromb Vasc Biol ; 26(12): 2696-702, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17008593

ABSTRACT

OBJECTIVE: Recent studies of bone marrow (BM)-transplanted apoE knockout (apoE-/-) mice have concluded that a substantial fraction of smooth muscle cells (SMCs) in atherosclerosis arise from circulating progenitor cells of hematopoietic origin. This pathway, however, remains controversial. In the present study, we reexamined the origin of plaque SMCs in apoE-/- mice by a series of BM transplantations and in a novel model of atherosclerosis induced in surgically transferred arterial segments. METHODS AND RESULTS: We analyzed plaques in lethally irradiated apoE-/- mice reconstituted with sex-mismatched BM cells from eGFP+ apoE-/- mice, which ubiquitously express enhanced green fluorescent protein (eGFP), but did not find a single SMC of donor BM origin among approximately 10,000 SMC profiles analyzed. We then transplanted arterial segments between eGFP+ apoE-/- and apoE-/- mice (isotransplantation except for the eGFP transgene) and induced atherosclerosis focally within the graft by a recently invented collar technique. No eGFP+ SMCs were found in plaques that developed in apoE-/- artery segments grafted into eGFP+ apoE-/- mice. Concordantly, 96% of SMCs were eGFP+ in plaques induced in eGFP+ apoE-/- artery segments grafted into apoE-/- mice. CONCLUSIONS: These experiments show that SMCs in atherosclerotic plaques are exclusively derived from the local vessel wall in apoE-/- mice.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/pathology , Hematopoietic Stem Cells/pathology , Muscle, Smooth, Vascular/pathology , Tunica Intima/pathology , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Bone Marrow Transplantation/pathology , Cell Differentiation , Cell Movement , Cell Proliferation , Disease Models, Animal , Female , Green Fluorescent Proteins , Hematopoietic Stem Cells/metabolism , Hyperlipidemias/pathology , Male , Mice , Mice, Knockout , Microscopy, Fluorescence/methods , Muscle, Smooth, Vascular/metabolism , Tunica Intima/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...