Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Cardiovasc Med ; 9: 971321, 2022.
Article in English | MEDLINE | ID: mdl-36148060

ABSTRACT

Background: CXCL12/CXCR4 signaling is essential in cardiac development and repair, however, its contribution to aortic valve stenosis (AVS) remains unclear. In this study, we tested the role of endothelial CXCR4 on the development of AVS. Materials and methods: We generated CXCR4 endothelial cell-specific knockout mice (EC CXCR4 KO) by crossing CXCR4fl/fl mice with Tie2-Cre mice to study the role of endothelial cell CXCR4 in AVS. CXCR4fl/fl mice were used as controls. Echocardiography was used to assess the aortic valve and cardiac function. Heart samples containing the aortic valve were stained using Alizarin Red for detection of calcification. Masson's trichrome staining was used for the detection of fibrosis. The apex of the heart samples was stained with wheat germ agglutinin (WGA) to visualize ventricular hypertrophy. Results: Compared with the control group, the deletion of CXCR4 in endothelial cells led to significantly increased aortic valve peak velocity and aortic valve peak pressure gradient, with decreased aortic valve area and ejection fraction. EC CXCR4 KO mice also developed cardiac hypertrophy as evidenced by increased diastolic and systolic left ventricle posterior wall thickness (LVPW), cardiac myocyte size, and heart weight (HW) to body weight (BW) ratio. Our data also confirmed increased microcalcifications, interstitial fibrosis, and thickened valvular leaflets of the EC CXCR4 KO mice. Conclusion: The data collected throughout this study suggest the deletion of CXCR4 in endothelial cells is linked to the development of aortic valve stenosis and left ventricular hypertrophy. The statistically significant parameters measured indicate that endothelial cell CXCR4 plays an important role in aortic valve development and function. We have compiled compelling evidence that EC CXCR4 KO mice can be used as a novel model for AVS.

2.
Stem Cells Transl Med ; 7(1): 115-124, 2018 01.
Article in English | MEDLINE | ID: mdl-29119710

ABSTRACT

Diabetes is a risk factor for worse outcomes following acute myocardial infarction (AMI). In this study, we tested the hypothesis that SDF-1:CXCR4 expression is compromised in post-AMI in diabetes, and that reversal of this defect can reverse the adverse effects of diabetes. Mesenchymal stem cells (MSC) isolated from green fluorescent protein (GFP) transgenic mice (control MSC) were induced to overexpress stromal cell-derived factor-1 (SDF-1). SDF-1 expression in control MSC and SDF-1-overexpressing MSC (SDF-1:MSC) were quantified using enzyme-linked immunosorbent assay (ELISA). AMI was induced on db/db and control mice. Mice were randomly selected to receive infusion of control MSC, SDF-1:MSC, or saline into the border zone after AMI. Serial echocardiography was used to assess cardiac function. SDF-1 and CXCR4 mRNA expression in the infarct zone of db/db mice and control mice were quantified. Compared to control mice, SDF-1 levels were decreased 82%, 91%, and 45% at baseline, 1 day and 3 days post-AMI in db/db mice, respectively. CXCR4 levels are increased 233% at baseline and 54% 5 days post-AMI in db/db mice. Administration of control MSC led to a significant improvement in ejection fraction (EF) in control mice but not in db/db mice 21 days after AMI. In contrast, administration of SDF-1:MSC produced a significant improvement in EF in both control mice and db/db mice 21 days after AMI. The SDF-1:CXCR4 axis is compromised in diabetes, which appears to augment the deleterious consequences of AMI. Over-express of SDF-1 expression in diabetes rescues cardiac function post AMI. Our results suggest that modulation of SDF-1 may improve post-AMI cardiac repair in diabetes. Stem Cells Translational Medicine 2018;7:115-124.


Subject(s)
Chemokine CXCL12/metabolism , Diabetes Mellitus/pathology , Mesenchymal Stem Cell Transplantation , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Receptors, CXCR4/metabolism , Animals , Apoptosis/physiology , Chemokine CXCL12/genetics , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Receptors, CXCR4/genetics , Stroke Volume/drug effects
3.
Stem Cells Transl Med ; 6(9): 1759-1766, 2017 09.
Article in English | MEDLINE | ID: mdl-28688176

ABSTRACT

Transplantation of adult stem cells into myocardial tissue after acute myocardial infarction (AMI), has been shown to improve tissue recovery and prevent progression to ischemic cardiomyopathy. Studies suggest that the effects of mesenchymal stem cells (MSC) are due to paracrine factors released by MSC, as the benefits of MSC can be achieved through delivery of conditioned media (CM) alone. We previously demonstrated that downregulation of Dab2 enhances MSC cardiac protein expression and improves cardiac function after AMI following MSC engraftment. In order to define the molecular mechanisms that regulate MSC secretome, we analyzed gene arrays in MSC following downregulation of Dab2 via TGFß1 pretreatment or transfection with Dab2:siRNA or miR-145. We identified 23 genes whose expressions were significantly changed in all three conditions. Among these genes, we have initially focused our validation and functional work on calcium/calmodulin-dependent protein kinase kinase-1 (CAMKK1). We quantified the effects of CAMKK1 overexpression in MSC following injection of CM after AMI. Injections of CM from MSC with CAMKK1 over-expression correlated with an increase in vascular density (CAMKK1 CM: 2,794.95 ± 44.2 versus Control: 1,290.69 ± 2.8 vessels/mm2 ) and decreased scar formation (CAMKK1 CM 50% ± 3.2% versus Control: 28% ± 1.4%), as well as improved cardiac function. Direct overexpression of CAMKK1 in infarcted tissue using a CAMKK1-encoding plasmid significantly improved ejection fraction (CAMKK1: 83.2% ± 5.4% versus saline: 51.7% ± 5.8%. Baseline: 91.3% ± 4.3%) and decreased infarct size after AMI. Our data identify a novel role for CAMKK1 as regulator of the MSC secretome and demonstrate that direct overexpression of CAMKK1 in infarcted cardiac tissue, results in therapeutic beneficial effects. Stem Cells Translational Medicine 2017;6:1759-1766.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Mesenchymal Stem Cells/metabolism , Proteome/metabolism , Regeneration , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cells, Cultured , Culture Media, Conditioned/pharmacology , Heart/drug effects , Heart/physiology , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Proteome/genetics , Rats , Rats, Inbred Lew
4.
Am J Physiol Heart Circ Physiol ; 310(1): H20-8, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26519029

ABSTRACT

The stromal cell-derived factor-1 (SDF-1):CXCR4 is important in myocardial repair. In this study we tested the hypothesis that early upregulation of cardiomyocyte CXCR4 (CM-CXCR4) at a time of high myocardial SDF-1 expression could be a strategy to engage the SDF-1:CXCR4 axis and improve cardiac repair. The effects of the hypoxia inducible factor (HIF) hydroxylase inhibitor dimethyloxalylglycine (DMOG) on CXCR4 expression was tested on H9c2 cells. In mice a myocardial infarction (MI) was produced in CM-CXCR4 null and wild-type controls. Mice were randomized to receive injection of DMOG (DMOG group) or saline (Saline group) into the border zone after MI. Protein and mRNA expression of CM-CXCR4 were quantified. Echocardiography was used to assess cardiac function. During hypoxia, DMOG treatment increased CXCR4 expression of H9c2 cells by 29 and 42% at 15 and 24 h, respectively. In vivo DMOG treatment increased CM-CXCR4 expression at 15 h post-MI in control mice but not in CM-CXCR4 null mice. DMOG resulted in increased ejection fraction in control mice but not in CM-CXCR4 null mice 21 days after MI. Consistent with greater cardiomyocyte survival with DMOG treatment, we observed a significant increase in cardiac myosin-positive area within the infarct zone after DMOG treatment in control mice, but no increase in CM-CXCR4 null mice. Inhibition of cardiomyocyte death in MI through the stabilization of HIF-1α requires downstream CM-CXCR4 expression. These data suggest that engagement of the SDF-1:CXCR4 axis through the early upregulation of CM-CXCR4 is a strategy for improving cardiac repair after MI.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Cardiotonic Agents/pharmacology , Myocardial Infarction/drug therapy , Myocardium/metabolism , Receptors, CXCR4/metabolism , Ventricular Function, Left/drug effects , Animals , Apoptosis/drug effects , Cell Hypoxia , Cell Line , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Rats , Receptors, CXCR4/deficiency , Receptors, CXCR4/genetics , Recovery of Function , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Stroke Volume/drug effects , Time Factors , Up-Regulation
5.
Am J Physiol Renal Physiol ; 308(2): F92-F100, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25377914

ABSTRACT

Childbirth injures muscles and nerves responsible for urinary continence. Mesenchymal stem cells (MSCs) or their secretome given systemically could provide therapeutic benefit for this complex multisite injury. We investigated whether MSCs or their secretome, as collected from cell culture, facilitate recovery from simulated childbirth injury. Age-matched female Sprague-Dawley rats received pudendal nerve crush and vaginal distension (PNC+VD) and a single intravenous (iv) injection of 2 million MSCs or saline. Controls received sham injury and iv saline. Additional rats received PNC+VD and a single intraperitoneal (ip) injection of concentrated media conditioned by MSCs (CCM) or concentrated control media (CM). Controls received a sham injury and ip CM. Urethral and nerve function were assessed with leak point pressure (LPP) and pudendal nerve sensory branch potential (PNSBP) recordings 3 wk after injury. Urethral and pudendal nerve anatomy were assessed qualitatively by blinded investigators. Quantitative data were analyzed using one-way ANOVA and Holm-Sidak post hoc tests with P < 0.05 indicating significant differences. Both LPP and PNSBP were significantly decreased 3 wk after PNC+VD with saline or CM compared with sham-injured rats, but not with MSC or CCM. Elastic fiber density in the urethra increased and changed in orientation after PNC+VD, with a greater increase in elastic fibers with MSC or CCM. Pudendal nerve fascicles were less dense and irregularly shaped after PNC+VD and had reduced pathology with MSC or CCM. MSC and CCM provide similar protective effects after PNC+VD, suggesting that MSCs act via their secretions in this dual muscle and nerve injury.


Subject(s)
Mesenchymal Stem Cell Transplantation , Pudendal Nerve/physiology , Urethra/physiology , Urinary Incontinence, Stress/prevention & control , Animals , Culture Media, Conditioned , Female , Injections, Intraperitoneal , Injections, Intravenous , Mesenchymal Stem Cells/metabolism , Parturition , Pudendal Nerve/injuries , Rats, Sprague-Dawley , Urethra/injuries , Urinary Incontinence, Stress/etiology
6.
Cell Transplant ; 23(11): 1395-406, 2014.
Article in English | MEDLINE | ID: mdl-23866688

ABSTRACT

Vaginal delivery is a risk factor for stress urinary incontinence (SUI). Mesenchymal stem cells (MSCs) home to injured organs and can facilitate repair. The goal of this study was to determine if MSCs home to pelvic organs after simulated childbirth injury and facilitate recovery from SUI via paracrine factors. Three experiments were performed. Eighteen female rats received vaginal distension (VD) or sham VD and labeled intravenous (IV) MSCs to investigate if MSCs home to the pelvic organs. Whole-organ imaging and immunofluorescence were performed 1 week later. Thirty-four female rats received VD and IV MSCs, VD and IV saline, or sham VD and IV saline to investigate if MSCs accelerate recovery of continence. Twenty-nine female rats received VD and periurethral concentrated conditioned media (CCM), VD and periurethral control media, or sham VD and periurethral control media to investigate if factors secreted by MSCs accelerate recovery from VD. Urethral histology and function were assessed 1 week later. Significantly more MSCs were observed in the urethra, vagina, and spleen after VD compared to sham VD. Continence as measured by leak point pressure (LPP) was significantly reduced after VD in rats treated with saline or control media compared to sham VD but not in those given MSCs or CCM. External urethral sphincter (EUS) function as measured by electromyography (EMG) was not improved with MSCs or CCM. Rats treated with MSCs or CCM demonstrated an increase in elastin fibers near the EUS and urethral smooth muscle more similar to that of sham-injured animals than rats treated with saline or control media. MSCs homed to the urethra and vagina and facilitated recovery of continence most likely via secretion of paracrine factors. Both MSCs and CCM have promise as novel noninvasive therapies for SUI.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Urinary Incontinence, Stress/therapy , Vagina/injuries , Animals , Delivery, Obstetric/adverse effects , Disease Models, Animal , Electromyography , Female , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Parturition , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley , Urinary Incontinence, Stress/etiology , Vagina/pathology
7.
PLoS One ; 8(7): e68528, 2013.
Article in English | MEDLINE | ID: mdl-23874657

ABSTRACT

RATIONALE: Stage specific embryonic antigen 1+ (SSEA1+) cells have been described as the most primitive mesenchymal progenitor cell in the bone marrow. Cardiac injury mobilizes SSEA1+ cells into the peripheral blood but their in vivo function has not been characterized. OBJECTIVE: We generated animals with chimeric bone marrow to determine the fate and function of bone marrow SSEA1+ cells in response to acute cardiac pressure overload. METHODS AND RESULTS: Lethally irradiated mice were transplanted with normal bone marrow where the wild-type SSEA1+ cells were replaced with green fluorescent protein (GFP) SSEA1+ cells. Cardiac injury was induced by trans-aortic constriction (TAC). We identified significant GFP+ cell engraftment into the myocardium after TAC. Bone marrow GFP+ SSEA1 derived cells acquired markers of endothelial lineage, but did not express markers of c-kit+ cardiac progenitor cells. The function of bone marrow SSEA1+ cells after TAC was determined by transplanting lethally irradiated mice with bone marrow depleted of SSEA1+ cells (SSEA1-BM). The cardiac function of SSEA1-BM mice declined at a greater rate after TAC compared to their complete bone marrow transplant counterparts and was associated with decreased bone marrow cell engraftment and greater vessel rarefication in the myocardium. CONCLUSIONS: These results provide evidence for the recruitment of endogenous bone marrow SSEA1+ cells to the myocardium after TAC. We demonstrate that, in vivo, bone marrow SSEA1+ cells have the differentiation potential to acquire endothelial lineage markers. We also show that bone marrow SSEA1+ deficiency is associated with a reduced compensatory capacity to cardiac pressure overload, suggesting their importance in cardiac homeostasis. These data demonstrate that bone marrow SSEA1+ cells are critical for sustaining vascular density and cardiac repair to pressure overload.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Lewis X Antigen/metabolism , Myocardium/metabolism , Animals , Bone Marrow Cells/cytology , Cell Tracking , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Ventricular Remodeling
8.
Cytotherapy ; 14(8): 983-93, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22624699

ABSTRACT

BACKGROUND AIMS: Acute cardiac injury results in the activation and recruitment of resident and non-cardiac stem cells. In this study we sought to define the pattern of peripheral stem cells and resident cardiac stem cell (CSC) activation induced acutely by cardiac pressure overload (PO). METHODS: PO was induced in mice by transaortic constriction (TAC). CSC, endothelial progenitor cells (EPC), hematopoietic stem cells (HSC) and stage-specific embryonic antigen (SSEA)-1(+) cells were profiled in the heart, spleen and bone marrow after TAC by flow cytometry. RESULTS: The combination of a systemic and local stem cell response resulted in increases in SSEA-1 (+) cells and EPC in the heart 7 and 14 days post-TAC, respectively. Locally, modest SSEA-1(+) proliferation at 4 days preceded the elevated myocardial stem cell number. We observed no significant proliferation of EPC and CSC in the heart. The systemic stem cell response was characterized by a biphasic loss of splenic SSEA-1(+) cells at 2 and 7 days post-TAC and loss of bone marrow and spleen EPC at 4 and 7 days, respectively. Spleen size changed dynamically after TAC. A negligible response of HSC to TAC was observed. Significant EPC and SSEA-1(+) proliferation in the bone marrow and spleen occurred only after their local levels were decreased. CONCLUSIONS: Our results demonstrate that an orchestrated systemic stem cell response (EPC and SSEA-1 (+) ) takes place in response to TAC. The increase of SSEA-1(+) cells and EPC in the heart in response to pressure is likely to be because of a combination of local proliferation and stem cell recruitment.


Subject(s)
Bone Marrow , Endothelial Cells , Hematopoietic Stem Cells , Myocardium , Spleen , Animals , Bone Marrow/metabolism , Cell Proliferation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Heart Injuries , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lewis X Antigen/metabolism , Mice , Myocardium/metabolism , Pressure , Spleen/metabolism
9.
Obstet Gynecol Int ; 2012: 612946, 2012.
Article in English | MEDLINE | ID: mdl-21941558

ABSTRACT

The local route of stem cell administration utilized presently in clinical trials for stress incontinence may not take full advantage of the capabilities of these cells. The goal of this study was to evaluate if intravenously injected mesenchymal stem cells (MSCs) home to pelvic organs after simulated childbirth injury in a rat model. Female rats underwent either vaginal distension (VD) or sham VD. All rats received 2 million GFP-labeled MSCs intravenously 1 hour after injury. Four or 10 days later pelvic organs and muscles were imaged for visualization of GFP-positive cells. Significantly more MSCs home to the urethra, vagina, rectum, and levator ani muscle 4 days after VD than after sham VD. MSCs were present 10 days after injection but GFP intensity had decreased. This study provides basic science evidence that intravenous administration of MSCs could provide an effective route for cell-based therapy to facilitate repair after injury and treat stress incontinence.

10.
J Orthop Res ; 29(7): 1064-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21567452

ABSTRACT

Homing of osteogenic cells through the systemic circulation represents an alternative to traditional orthopedic tissue engineering approaches that focus on local cell populations. We hypothesize that expression of the chemokine, stromal cell-derived factor-1 (SDF-1) or monocyte chemotactic protein-3 (MCP-3) may enhance homing of osteogenic cells into sites of fracture repair, as both have demonstrated promise in recruitment of marrow stromal cells (MSCs). This hypothesis was tested by transplantation of culture expanded MSCs expressing these factors adjacent to a fracture site on a collagen scaffold. One green fluorescent protein positive (GFP+) and one wild-type mouse were surgically conjoined as parabiots at 7-8 weeks of age. Fibular osteotomy was performed 4 weeks after parabiosis on the hind limb of the wild-type mouse. Mice were randomly allocated to receive one of the following five treatments: control (no scaffold), empty scaffold (no cells), or scaffold containing MSCs, scaffold containing MSCs expressing SDF-1, or scaffold containing MSCs expressing MCP-3. Fracture callus was harvested 2 weeks after injury, and analyzed with confocal microscopy and cell-counting software. When compared to fracture callus treated with nontransfected MSCs, the fracture callus of mice treated with both SDF-1 and MCP-3 secreting MSCs demonstrated a significant increase in the number of both GFP+ cells (p = 0.0003, p = 0.02) and GFP+ /AP+ cells (p = 0.0005, p = 0.01). These data suggest that homing of osteogenic cells from systemic circulation participate in fracture repair and that homing pathways might be modulated to enhance the contribution of circulating progenitors at the site of skeletal injury.


Subject(s)
Chemokine CCL7/genetics , Chemokine CXCL12/genetics , Fracture Healing/physiology , Fractures, Bone/physiopathology , Osteocytes/physiology , Animals , Cell Movement/physiology , Chemokine CCL7/metabolism , Chemokine CXCL12/metabolism , Female , Fractures, Bone/metabolism , Fractures, Bone/surgery , Green Fluorescent Proteins/genetics , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteocytes/cytology
11.
FASEB J ; 21(12): 3197-207, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17496162

ABSTRACT

Stem cell transplantation at the time of acute myocardial infarction (AMI) improves cardiac function. Whether the improved cardiac function results from regeneration of cardiac myocytes, modulation of remodeling, or preservation of injured tissue through paracrine mechanisms is actively debated. Because no specific stem cell population has been shown to be optimal, we investigated whether the benefit of stem cell transplantation could be attributed to a trophic effect on injured myocardium. Mesenchymal stem cells secrete SDF-1 and the interaction of SDF-1 with its receptor, CXCR4, increases survival of progenitor cells. Therefore, we compared the effects of MSC and MSC engineered to overexpress SDF-1 on cardiac function after AMI. Tail vein infusion of syngeneic MSC and MSC:SDF-1 1 day after AMI in the Lewis rat led to improved cardiac function by echocardiography by 70.7% and 238.8%, respectively, compared with saline controls 5 wk later. The beneficial effects of MSC and MSC:SDF-1 transplantation were mediated primarily through preservation, not regeneration of cardiac myocytes within the infarct zone. The direct effect of SDF-1 on cardiac myocytes was due to the observation that, between 24 and 48 h after AMI, SDF-1-expressing MSC increased cardiac myocyte survival, vascular density (18.2+/-4.0 vs. 7.6+/-2.3 vessels/mm2, P<0.01; SDF-1:MSC vs. MSC), and cardiac myosin-positive area (MSC: 49.5%; mSC:SDF-1: 162.1%) within the infarct zone. There was no evidence of cardiac regeneration by the infused MSC or endogenous cardiac stem cells based on lack of evidence for cardiac myocytes being derived from replicating cells. These results indicate that stem cell transplantation may have significant beneficial effects on injured organ function independent of tissue regeneration and identify SDF-1:CXCR4 binding as a novel target for myocardial preservation.


Subject(s)
Chemokine CXCL12/metabolism , Mesenchymal Stem Cells/physiology , Myocardial Infarction , Myocytes, Cardiac/metabolism , Stem Cell Transplantation , Animals , Biomarkers/metabolism , Cell Survival , Cells, Cultured , Chemokine CXCL12/genetics , Fluorescent Dyes/metabolism , Hypoxia , Mesenchymal Stem Cells/cytology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardial Ischemia , Myocytes, Cardiac/cytology , Rats , Rats, Inbred Lew , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
12.
Circ Res ; 100(11): 1626-33, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17495229

ABSTRACT

Recent loss-of-function studies highlight the importance of the transcription factor GATA4 in the myocardial response to injury in the adult heart. However, the potential effects of gain-in-function of GATA4 overexpression, and transcription factors in general, is hindered by the fact that transcription factors are neither secreted nor taken up by cells. Chimeric proteins incorporating motifs of cell-penetrating proteins are secreted from cells and internalized by surrounding cells. We engineered a chimeric protein consisting of GATA4 and the cell-penetrating protein VP22. Cardiac fibroblasts stably transfected with the GATA4:VP22, GFP:VP22, or green fluorescent protein (GFP) constructs were transplanted into Lewis rats 1 month after left anterior descending ligation. GATA4:VP22 expression in the border zone was associated with increased cardiac myosin expression and cardiac myocyte size (30 mum versus 19 mum, P<0.01). Compared with the GFP:VP22 control group, 6 weeks after cardiac fibroblast transplantation (10 weeks after myocardial infarction), animals that received GATA4:VP22-expressing cardiac fibroblasts demonstrated increased cardiac function and less negative remodeling. These data demonstrate a novel strategy for transcription factor delivery to injured myocardium and indicate that the delivery of GATA4 locally to the infarct border zone induces multiple local effects in the border zone cardiac myocytes resulting in beneficial ventricular remodeling and improved global left ventricular function.


Subject(s)
Fibroblasts/metabolism , GATA4 Transcription Factor/genetics , Genetic Therapy/methods , Myocardial Infarction/therapy , Myocardial Ischemia/therapy , Recombinant Fusion Proteins/genetics , Animals , Cell Size , Cells, Cultured , Disease Models, Animal , Fibroblasts/transplantation , GATA4 Transcription Factor/metabolism , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/biosynthesis , Transfection , Ventricular Function, Left/genetics , Viral Structural Proteins/genetics
13.
J Mol Cell Cardiol ; 42(2): 304-14, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17070540

ABSTRACT

Clinical studies suggest increased arrhythmia risk associated with cell therapy for myocardial infarction (MI); however, the underlying mechanisms are poorly understood. We hypothesize that the degree of electrical viability in the infarct and border zone associated with skeletal myoblast (SKMB) or mesenchymal stem cell (MSC) therapy will determine arrhythmia vulnerability in the whole heart. Within 24 h of LAD ligation in rats, 2 million intramyocardially injected SKMB (n=6), intravenously infused MSC (n=7), or saline (n=7) was administered. One month after MI, cardiac function was determined and novel optical mapping techniques were used to assess electrical viability and arrhythmia inducibility. Shortening fraction was greater in rats receiving SKMB (17.8%+/-5.3%, p=0.05) or MSC (17.6%+/-3.0%, p<0.01) compared to MI alone (10.1%+/-2.2%). Arrhythmia inducibility score was significantly greater in SKMB (2.8+/-0.2) compared to MI (1.4+/-0.5, p=0.05). Inducibility score for MSC (0.6+/-0.4) was significantly lower than SKMB (p=0.01) and tended to be lower than MI. Optical mapping revealed that MSC therapy preserved electrical viability and impulse propagation in the border zone, but SKMB did not. In addition, injected SKMBs were localized to discrete cell clusters where connexin expression was absent. In contrast, infused MSCs engrafted in a more homogeneous pattern and expressed connexin proteins. Even though both MSC and SKMB therapy improved cardiac function following MI in rat, SKMB therapy significantly increased arrhythmia inducibility while MSC therapy tended to lower inducibility. In addition, only MSC therapy was associated with enhanced electrical viability, diffuse engraftment, and connexin expression, which may explain the differences in arrhythmia inducibility.


Subject(s)
Mesenchymal Stem Cell Transplantation , Myoblasts, Skeletal/transplantation , Myocardial Infarction/therapy , Recovery of Function , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/therapy , Cell Survival , Electrophysiologic Techniques, Cardiac , Graft Survival , Heart Conduction System/pathology , Heart Conduction System/physiopathology , Myoblasts, Skeletal/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats , Transplantation, Homologous
14.
Lancet ; 362(9385): 697-703, 2003 Aug 30.
Article in English | MEDLINE | ID: mdl-12957092

ABSTRACT

BACKGROUND: Myocardial regeneration via stem-cell mobilisation at the time of myocardial infarction is known to occur, although the mechanism for stem-cell homing to infarcted tissue subsequently and whether this approach can be used for treatment of ischaemic cardiomyopathy are unknown. We investigated these issues in a Lewis rat model (ligation of the left anterior descending artery) of ischaemic cardiomyopathy. METHODS: We studied the effects of stem-cell mobilisation by use of granulocyte colony-stimulating factor (filgrastim) with or without transplantation of syngeneic cells. Shortening fraction and myocardial strain by tissue doppler imaging were quantified by echocardiography. FINDINGS: Stem-cell mobilisation with filgrastim alone did not lead to engraftment of bone-marrow-derived cells. Stromal-cell-derived factor 1 (SDF-1), required for stem-cell homing to bone marrow, was upregulated immediately after myocardial infarction and downregulated within 7 days. 8 weeks after myocardial infarction, transplantation into the peri-infarct zone of syngeneic cardiac fibroblasts stably transfected to express SDF-1 induced homing of CD117-positive stem cells to injured myocardium after filgrastim administration (control vs SDF-1-expressing cardiac fibroblasts mean 7.2 [SD 3.4] vs 33.2 [6.0] cells/mm2, n=4 per group, p<0.02) resulting in greater left-ventricular mass (1.24 [0.29] vs 1.57 [0.27] g) and better cardiac function (shortening fraction 9.2 [4.9] vs 17.2 [4.2]%, n=8 per group, p<0.05). INTERPRETATION: These findings show that SDF-1 is sufficient to induce therapeutic stem-cell homing to injured myocardium and suggest a strategy for directed stem-cell engraftment into injured tissues. Our findings also indicate that therapeutic strategies focused on stem-cell mobilisation for regeneration of myocardial tissue must be initiated within days of myocardial infarction unless signalling for stem-cell homing is re-established.


Subject(s)
Cell Movement/physiology , Chemokines, CXC/physiology , Myocardial Ischemia/surgery , Pluripotent Stem Cells/physiology , Regeneration/physiology , Stem Cell Transplantation/methods , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Chemokine CXCL12 , Colony-Stimulating Factors/administration & dosage , Colony-Stimulating Factors/pharmacology , Disease Models, Animal , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/pharmacology , Pluripotent Stem Cells/drug effects , Rats , Rats, Inbred Lew , Recombinant Proteins , Regeneration/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...