Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747287

ABSTRACT

Lymphedema is a debilitating disease with no effective cure and affects an estimated 250 million individuals worldwide. Prior studies have identified mutations in piezo-type mechanosensitive ion channel component 1 (PIEZO1), angiopoietin 2 (ANGPT2), and tyrosine kinase with Ig-like and EGF-like domains 1 (TIE1) in patients with primary lymphedema. Here, we identified crosstalk between these molecules and showed that activation of the mechanosensory channel PIEZO1 in lymphatic endothelial cells (LECs) caused rapid exocytosis of the TIE ligand ANGPT2, ectodomain shedding of TIE1 by disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), and increased TIE/PI3K/AKT signaling, followed by nuclear export of the transcription factor FOXO1. These data establish a functional network between lymphedema-associated genes and provide what we believe to be the first molecular mechanism bridging channel function with vascular signaling and intracellular events culminating in transcriptional regulation of genes expressed in LECs. Our study provides insights into the regulation of lymphatic function and molecular pathways involved in human disease.


Subject(s)
Angiopoietin-2 , Forkhead Box Protein O1 , Ion Channels , Lymphangiogenesis , Lymphedema , Receptor, TIE-1 , Signal Transduction , Animals , Humans , Mice , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Angiopoietin-2/metabolism , Angiopoietin-2/genetics , Endothelial Cells/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Ion Channels/metabolism , Ion Channels/genetics , Lymphangiogenesis/genetics , Lymphedema/metabolism , Lymphedema/genetics , Lymphedema/pathology , Mechanotransduction, Cellular , Receptor, TIE-1/metabolism , Receptor, TIE-1/genetics
2.
Nat Commun ; 12(1): 6072, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663817

ABSTRACT

Primary congenital glaucoma (PCG) is a severe disease characterized by developmental defects in the trabecular meshwork (TM) and Schlemm's canal (SC), comprising the conventional aqueous humor outflow pathway of the eye. Recently, heterozygous loss of function variants in TEK and ANGPT1 or compound variants in TEK/SVEP1 were identified in children with PCG. Moreover, common variants in ANGPT1and SVEP1 have been identified as risk alleles for primary open angle glaucoma (POAG) in GWAS studies. Here, we show tissue-specific deletion of Angpt1 or Svep1 from the TM causes PCG in mice with severe defects in the adjacent SC. Single-cell transcriptomic analysis of normal and glaucomatous Angpt1 deficient eyes allowed us to identify distinct TM and SC cell populations and discover additional TM-SC signaling pathways. Furthermore, confirming the importance of angiopoietin signaling in SC, delivery of a recombinant ANGPT1-mimetic promotes developmental SC expansion in healthy and Angpt1 deficient eyes, blunts intraocular pressure (IOP) elevation and RGC loss in a mouse model of PCG and lowers IOP in healthy adult mice. Our data highlight the central role of ANGPT1-TEK signaling and TM-SC crosstalk in IOP homeostasis and provide new candidates for SC-targeted glaucoma therapy.


Subject(s)
Aqueous Humor/metabolism , Cell Communication/physiology , Glaucoma, Open-Angle/pathology , Glaucoma, Open-Angle/therapy , Angiopoietin-1/administration & dosage , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Anterior Chamber/blood supply , Anterior Chamber/cytology , Anterior Chamber/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Communication/drug effects , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Profiling , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/metabolism , Intraocular Pressure/drug effects , Intraocular Pressure/genetics , Mice , Mice, Knockout , Neural Crest/cytology , Neural Crest/metabolism , Proteins/genetics , Proteins/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Single-Cell Analysis , Trabecular Meshwork/cytology , Trabecular Meshwork/metabolism
3.
JCI Insight ; 5(17)2020 09 03.
Article in English | MEDLINE | ID: mdl-32879134

ABSTRACT

Diabetes is a risk factor for myocardial infarction, and outcomes after myocardial infarction are worse among diabetics compared with nondiabetics. Diabetes is associated with impaired Heme clearance. Here, we determined whether heme toxicity and impaired heme clearance contribute to diabetic myocardial infarction injury and assessed IL-10 as a therapeutic agent for diabetic myocardial infarction. Plasma-free hemoglobin was significantly elevated in diabetic mice compared with nondiabetic mice after myocardial infarction. Infarct size had strong correlation to the level of plasma-free hemoglobin. Hemoglobin and reactive iron deposition within the infarct zone were also demonstrated in diabetic MI. IL-10 significantly reduced infarct size and improved cardiac function in diabetic mice. Moreover, IL-10 improved capillary density, reduced apoptosis, and decreased inflammation in the border zone of the infarcted hearts, findings that were partially inhibited by Tin protoporphyrin (a heme oxygenase-1 inhibitor). IL-10 upregulated CD163, the hemoglobin:haptoglobin scavenger receptor, and heme oxygenase-1 in THP-1-derived and primary human CD14+ macrophages. IL-10 significantly protected against ischemic injury when HL-1 cardiomyocytes were cotreated with hemoglobin. Together, our findings indicate that IL-10 is cardioprotective in diabetic myocardial infarction via upregulation of heme clearance pathways. These findings implicate heme clearance as a potentially novel therapeutic direction for diabetic myocardial infarction.


Subject(s)
Diabetic Cardiomyopathies/drug therapy , Heme/metabolism , Interleukin-10/therapeutic use , Myocardial Infarction/drug therapy , Animals , Apoptosis , Cells, Cultured , Diabetic Cardiomyopathies/metabolism , Hemoglobins/metabolism , Humans , Interleukin-10/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , THP-1 Cells
4.
NMR Biomed ; 33(9): e4327, 2020 09.
Article in English | MEDLINE | ID: mdl-32567177

ABSTRACT

BACKGROUND: Doxorubicin and doxorubicin-trastuzumab combination chemotherapy have been associated with cardiotoxicity that eventually leads to heart failure and may limit dose-effective cancer treatment. Current diagnostic strategies rely on decreased ejection fraction (EF) to diagnose cardiotoxicity. PURPOSE: The aim of this study is to explore the potential of cardiac MR (CMR) imaging to identify imaging biomarkers in a mouse model of chemotherapy-induced cardiotoxicity. METHODS: A cumulative dose of 25 mg/kg doxorubicin was administered over three weeks using subcutaneous pellets (n = 9, Dox). Another group (n = 9) received same dose of Dox and a total of 10 mg/kg trastuzumab (DT). Mice were imaged at baseline, 5/6 weeks and 10 weeks post-treatment on a 7T MRI system. The protocol included short-axis cine MRI covering the left ventricle (LV) and mid-ventricular short-axis tissue phase mapping (TPM), pre- and post-contrast T1 mapping, T2 mapping and Displacement Encoding with Stimulated Echoes (DENSE) strain encoded MRI. EF, peak myocardial velocities, native T1, T2, extracellular volume (ECV), and myocardial strain were quantified. N = 7 mice were sacrificed for histopathologic assessment of apoptosis at 5/6 weeks. RESULTS: Global peak systolic longitudinal velocity was reduced at 5/6 weeks in Dox (0.6 ± 0.3 vs 0.9 ± 0.3, p = 0.02). In the Dox group, native T1 was reduced at 5/6 weeks (1.3 ± 0.2 ms vs 1.6 ± 0.2 ms, p = 0.02), and relatively normalized at week 10 (1.4 ± 0.1 ms vs 1.6 ± 0.2 ms, p > 0.99). There was no change in EF and other MRI parameters and histopathologic results demonstrated minimal apoptosis in all mice (~1-2 apoptotic cell/high power field), suggesting early-stage cardiotoxicity. CONCLUSIONS: In a mouse model of chemotherapy-induced cardiotoxicity using doxorubicin and trastuzumab, advanced CMR shows promise in identifying treatment-related decrease in myocardial velocity and native T1 prior to the onset of cardiomyocyte apoptosis and reduction of EF.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxicity/physiopathology , Heart/physiopathology , Magnetic Resonance Imaging , Animals , Body Weight , Disease Models, Animal , Doxorubicin/adverse effects , Hematocrit , Mice, Inbred C57BL , Myocardium/pathology , Myocardium/ultrastructure , Stroke Volume/physiology , Systole/physiology , Trastuzumab/adverse effects
5.
Cardiovasc Toxicol ; 19(5): 482-484, 2019 10.
Article in English | MEDLINE | ID: mdl-31028602

ABSTRACT

An increasing volume of pre-clinical and clinical-translational research is attempting to identify novel biomarkers for improved diagnosis and risk-stratification of chemotherapy-induced cardiotoxicity. Most published animal models have employed weekly intraperitoneal injections of doxorubicin to reach a desired cumulative dose. This approach can be associated with severe systemic toxicity which limits the animal model usefulness, particularly for advanced imaging. In the current study, slow-release subcutaneous doxorubicin pellets demonstrated histopathologic evidence of cardiotoxicity at doses similar to standard human dose-equivalents without limiting animal survival or ability to participate in advanced imaging studies. This approach may provide a more robust cardiotoxicity animal model.


Subject(s)
Antibiotics, Antineoplastic , Doxorubicin , Heart Diseases/chemically induced , Myocytes, Cardiac/pathology , Animals , Cardiotoxicity , Disease Models, Animal , Drug Implants , Female , Heart Diseases/diagnostic imaging , Heart Diseases/pathology , Mice, Inbred C57BL , Time Factors
6.
Lab Invest ; 98(5): 682-691, 2018 05.
Article in English | MEDLINE | ID: mdl-29453401

ABSTRACT

Sonic Hedgehog (Shh) signaling induces neovascularization and angiogenesis. It is not known whether the hedgehog signaling pathway in endothelial cells is essential to angiogenesis. Smoothened (Smo) transduces hedgehog signaling across the cell membrane. This study assessed whether endothelial Smoothened-dependent Shh signaling is required for Shh-mediated angiogenesis and ischemic tissue repair. Endothelial-specific smoothened knockout mice, eSmoNull were created using Cre-lox recombination system. eSmoNull mice had no observable phenotype at baseline and showed normal cardiac function. Smoothened in CD31+ cells isolated from eSmoNull hearts was significantly reduced compared to CD31+ cells from eSmoWT littermate control hearts. Fluorescence immunostaining of eSmoNull heart sections showed Smo expression in endothelial cells was abolished. The hind-limb ischemia (HLI) model was used to assess the response to ischemic injury. Perfusion ratio, limb motor function, and limb necrosis were not significantly different after HLI between eSmoNull mice and eSmoWT. Capillary densities in the ischemic limb in eSmoNull mice were also similar to eSmoWT at 4 weeks after HLI. Next, response to exogenous Shh was assessed in the corneal angiogenesis model. There was no significant difference in corneal angiogenesis induced by administration of Shh pellets between eSmoWT and eSmoNull mice. Furthermore, in vitro experiments demonstrated that direct Shh had limited effects on endothelial cell proliferation and migration. However, conditioned media from Shh-treated fibroblasts had a more potent effect on endothelial cell proliferation and migration than non-treated conditioned media. Furthermore, Shh treatment of fibroblasts dramatically stimulated angiogenic growth factor expression, including PDGF-B, VEGF-A, HGF and IGF. PDGF-B was the most upregulated and may contribute to the large neo-vessels associated with Shh-induced angiogenesis. Taken together, these data demonstrate that Shh signaling via Smoothened in endothelial cells is not required for angiogenesis and ischemic tissue repair. Shh signaling via stromal cells likely mediates its angiogenic effects.


Subject(s)
Endothelial Cells/physiology , Hedgehog Proteins/physiology , Ischemia/physiopathology , Neovascularization, Physiologic , Signal Transduction/physiology , Smoothened Receptor/physiology , Animals , Cells, Cultured , Fibroblasts/physiology , Hindlimb/blood supply , Male , Mice
7.
ACS Appl Mater Interfaces ; 10(8): 6904-6916, 2018 Feb 28.
Article in English | MEDLINE | ID: mdl-29385802

ABSTRACT

Disorders of blood vessels cause a range of severe health problems. As a powerful vasodilator and cellular second messenger, nitric oxide (NO) is known to have beneficial vascular functions. However, NO typically has a short half-life and is not specifically targeted. On the other hand, high-density lipoproteins (HDLs) are targeted natural nanoparticles (NPs) that transport cholesterol in the systemic circulation and whose protective effects in vascular homeostasis overlap with those of NO. Evolving the AuNP-templated HDL-like nanoparticles (HDL NPs), a platform of bioinspired HDL, we set up a targeted biomimetic nanotherapy for vascular disease that combines the functions of NO and HDL. A synthetic S-nitrosylated (SNO) phospholipid (1,2-dipalmitoyl-sn-glycero-3-phosphonitrosothioethanol) was synthesized and assembled with S-containing phospholipids and the principal protein of HDL, apolipoprotein A-I, to construct NO-delivering HDL-like particles (SNO HDL NPs). SNO HDL NPs self-assemble under mild conditions similar to natural processes, avoiding the complex postassembly modification needed for most synthetic NO-release nanoparticles. In vitro data demonstrate that the SNO HDL NPs merge the functional properties of NO and HDL into a targeted nanocarrier. Also, SNO HDL NPs were demonstrated to reduce ischemia/reperfusion injury in vivo in a mouse kidney transplant model and atherosclerotic plaque burden in a mouse model of atherosclerosis. Thus, the synthesis of SNO HDL NPs provides not only a bioinspired nanotherapy for vascular disease but also a foundation to construct diversified multifunctional platforms based on HDL NPs in the future.


Subject(s)
Nanoparticles , Animals , Atherosclerosis , Biomimetics , Lipoproteins, HDL , Mice , Nitric Oxide
8.
Mol Pharm ; 14(11): 4042-4051, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28933554

ABSTRACT

Cancer cells have altered metabolism and, in some cases, an increased demand for cholesterol. It is important to identify novel, rational treatments based on biology, and cellular cholesterol metabolism as a potential target for cancer is an innovative approach. Toward this end, we focused on diffuse large B-cell lymphoma (DLBCL) as a model because there is differential cholesterol biosynthesis driven by B-cell receptor (BCR) signaling in germinal center (GC) versus activated B-cell (ABC) DLBCL. To specifically target cellular cholesterol homeostasis, we employed high-density lipoprotein-like nanoparticles (HDL NP) that can generally reduce cellular cholesterol by targeting and blocking cholesterol uptake through the high-affinity HDL receptor, scavenger receptor type B-1 (SCARB1). As we previously reported, GC DLBCL are exquisitely sensitive to HDL NP as monotherapy, while ABC DLBCL are less sensitive. Herein, we report that enhanced BCR signaling and resultant de novo cholesterol synthesis in ABC DLBCL drastically reduces the ability of HDL NPs to reduce cellular cholesterol and induce cell death. Therefore, we combined HDL NP with the BCR signaling inhibitor ibrutinib and the SYK inhibitor R406. By targeting both cellular cholesterol uptake and BCR-associated de novo cholesterol synthesis, we achieved cellular cholesterol reduction and induced apoptosis in otherwise resistant ABC DLBCL cell lines. These results in lymphoma demonstrate that reduction of cellular cholesterol is a powerful mechanism to induce apoptosis. Cells rich in cholesterol require HDL NP therapy to reduce uptake and molecularly targeted agents that inhibit upstream pathways that stimulate de novo cholesterol synthesis, thus, providing a new paradigm for rationally targeting cholesterol metabolism as therapy for cancer.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/metabolism , Nanoparticles/chemistry , Receptors, Antigen, B-Cell/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cholesterol/metabolism , Humans , Lipoproteins, HDL/metabolism , Receptors, Lipoprotein/metabolism , Scavenger Receptors, Class B/metabolism , Signal Transduction/physiology
9.
Circ Res ; 121(8): 930-940, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28851810

ABSTRACT

RATIONALE: Clinical benefits of reperfusion after myocardial infarction are offset by maladaptive innate immune cell function, and therapeutic interventions are lacking. OBJECTIVE: We sought to test the significance of phagocytic clearance by resident and recruited phagocytes after myocardial ischemia reperfusion. METHODS AND RESULTS: In humans, we discovered that clinical reperfusion after myocardial infarction led to significant elevation of the soluble form of MerTK (myeloid-epithelial-reproductive tyrosine kinase; ie, soluble MER), a critical biomarker of compromised phagocytosis by innate macrophages. In reperfused mice, macrophage Mertk deficiency led to decreased cardiac wound debridement, increased infarct size, and depressed cardiac function, newly implicating MerTK in cardiac repair after myocardial ischemia reperfusion. More notably, Mertk(CR) mice, which are resistant to cleavage, showed significantly reduced infarct sizes and improved systolic function. In contrast to other cardiac phagocyte subsets, resident cardiac MHCIILOCCR2- (major histocompatibility complex II/C-C motif chemokine receptor type 2) macrophages expressed higher levels of MerTK and, when exposed to apoptotic cells, secreted proreparative cytokines, including transforming growth factor-ß. Mertk deficiency compromised the accumulation of MHCIILO phagocytes, and this was rescued in Mertk(CR) mice. Interestingly, blockade of CCR2-dependent monocyte infiltration into the heart reduced soluble MER levels post-ischemia reperfusion. CONCLUSIONS: Our data implicate monocyte-induced MerTK cleavage on proreparative MHCIILO cardiac macrophages as a novel contributor and therapeutic target of reperfusion injury.


Subject(s)
Macrophages/enzymology , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , ST Elevation Myocardial Infarction/enzymology , Animals , Apoptosis , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunity, Innate , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Monocytes/enzymology , Monocytes/immunology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/immunology , Myocardium/pathology , Phagocytosis , Phenotype , Proteolysis , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , ST Elevation Myocardial Infarction/immunology , ST Elevation Myocardial Infarction/pathology , ST Elevation Myocardial Infarction/physiopathology , Signal Transduction , Time Factors , c-Mer Tyrosine Kinase
10.
Circ Res ; 120(9): 1466-1476, 2017 Apr 28.
Article in English | MEDLINE | ID: mdl-28298297

ABSTRACT

RATIONALE: Paracrine secretions seem to mediate therapeutic effects of human CD34+ stem cells locally transplanted in patients with myocardial and critical limb ischemia and in animal models. Earlier, we had discovered that paracrine secretion from human CD34+ cells contains proangiogenic, membrane-bound nanovesicles called exosomes (CD34Exo). OBJECTIVE: Here, we investigated the mechanisms of CD34Exo-mediated ischemic tissue repair and therapeutic angiogenesis by studying their miRNA content and uptake. METHODS AND RESULTS: When injected into mouse ischemic hindlimb tissue, CD34Exo, but not the CD34Exo-depleted conditioned media, mimicked the beneficial activity of their parent cells by improving ischemic limb perfusion, capillary density, motor function, and their amputation. CD34Exo were found to be enriched with proangiogenic miRNAs such as miR-126-3p. Knocking down miR-126-3p from CD34Exo abolished their angiogenic activity and beneficial function both in vitro and in vivo. Interestingly, injection of CD34Exo increased miR-126-3p levels in mouse ischemic limb but did not affect the endogenous synthesis of miR-126-3p, suggesting a direct transfer of stable and functional exosomal miR-126-3p. miR-126-3p enhanced angiogenesis by suppressing the expression of its known target, SPRED1, simultaneously modulating the expression of genes involved in angiogenic pathways such as VEGF (vascular endothelial growth factor), ANG1 (angiopoietin 1), ANG2 (angiopoietin 2), MMP9 (matrix metallopeptidase 9), TSP1 (thrombospondin 1), etc. Interestingly, CD34Exo, when treated to ischemic hindlimbs, were most efficiently internalized by endothelial cells relative to smooth muscle cells and fibroblasts, demonstrating a direct role of stem cell-derived exosomes on mouse endothelium at the cellular level. CONCLUSIONS: Collectively, our results have demonstrated a novel mechanism by which cell-free CD34Exo mediates ischemic tissue repair via beneficial angiogenesis. Exosome-shuttled proangiogenic miRNAs may signify amplification of stem cell function and may explain the angiogenic and therapeutic benefits associated with CD34+ stem cell therapy.


Subject(s)
Angiogenic Proteins/metabolism , Antigens, CD34/metabolism , Endothelial Progenitor Cells/transplantation , Exosomes/transplantation , Ischemia/surgery , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Angiogenic Proteins/genetics , Animals , Biomarkers/metabolism , Cells, Cultured , Culture Media, Conditioned/metabolism , Disease Models, Animal , Endothelial Progenitor Cells/metabolism , Exosomes/metabolism , Gene Expression Regulation , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Motor Activity , Paracrine Communication , Phenotype , RNA Interference , Recovery of Function , Regional Blood Flow , Signal Transduction , Time Factors , Transfection
11.
Vasc Cell ; 7: 6, 2015.
Article in English | MEDLINE | ID: mdl-26161255

ABSTRACT

BACKGROUND: Multiple studies demonstrated pro-angiogenic effects of microRNA (miR)-27b. Its targets include Notch ligand Dll4, Sprouty (Spry)-2, PPARγ and Semaphorin (SEMA) 6A. miR-27 effects in the heart are context-dependent: although it is necessary for ventricular maturation, targeted overexpression in cardiomyocytes causes hypertrophy and dysfunction during development. Despite significant recent advances, therapeutic potential of miR-27b in cardiovascular disease and its effects in adult heart remain unexplored. Here, we assessed the therapeutic potential of miR-27b mimics and inhibitors in rodent models of ischemic disease and cancer. METHODS: We have used a number of models to demonstrate the effects of miR-27b mimicry and inhibition in vivo, including subcutaneous Matrigel plug assay, mouse models of hind limb ischemia and myocardial infarction and subcutaneous Lewis Lung carcinoma. RESULTS: Using mouse model of myocardial infarction due to the coronary artery ligation, we showed that miR-27b mimic had overall beneficial effects, including increased vascularization, decreased fibrosis and increased ejection fraction. In mouse model of critical limb ischemia, miR-27b mimic also improved tissue re-vascularization and perfusion. In both models, miR-27b mimic clearly decreased macrophage recruitment to the site of hypoxic injury. In contrast, miR-27b increased the recruitment of bone marrow derived cells to the neovasculature, as was shown using mice reconstituted with fluorescence-tagged bone marrow. These effects were due, at least in part, to the decreased expression of Dll4, PPARγ and IL10. In contrast, blocking miR-27b significantly decreased vascularization and reduced growth of subcutaneous tumors and decreased BMDCs recruitment to the tumor vasculature. CONCLUSIONS: Our study demonstrates the utility of manipulating miR-27b levels in the treatment of cardiovascular disease and cancer.

12.
Circ Res ; 117(1): 52-64, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25904597

ABSTRACT

RATIONALE: Embryonic stem cells (ESCs) hold great promise for cardiac regeneration but are susceptible to various concerns. Recently, salutary effects of stem cells have been connected to exosome secretion. ESCs have the ability to produce exosomes, however, their effect in the context of the heart is unknown. OBJECTIVE: Determine the effect of ESC-derived exosome for the repair of ischemic myocardium and whether c-kit(+) cardiac progenitor cells (CPCs) function can be enhanced with ESC exosomes. METHODS AND RESULTS: This study demonstrates that mouse ESC-derived exosomes (mES Ex) possess ability to augment function in infarcted hearts. mES Ex enhanced neovascularization, cardiomyocyte survival, and reduced fibrosis post infarction consistent with resurgence of cardiac proliferative response. Importantly, mES Ex augmented CPC survival, proliferation, and cardiac commitment concurrent with increased c-kit(+) CPCs in vivo 8 weeks after in vivo transfer along with formation of bonafide new cardiomyocytes in the ischemic heart. miRNA array revealed significant enrichment of miR290-295 cluster and particularly miR-294 in ESC exosomes. The underlying basis for the beneficial effect of mES Ex was tied to delivery of ESC specific miR-294 to CPCs promoting increased survival, cell cycle progression, and proliferation. CONCLUSIONS: mES Ex provide a novel cell-free system that uses the immense regenerative power of ES cells while avoiding the risks associated with direct ES or ES-derived cell transplantation and risk of teratomas. ESC exosomes possess cardiac regeneration ability and modulate both cardiomyocyte and CPC-based repair programs in the heart.


Subject(s)
Embryonic Stem Cells/physiology , Exosomes/physiology , Myocardial Infarction/therapy , Animals , Cell Survival , Cell-Free System , Collagen , Drug Combinations , Embryonic Stem Cells/ultrastructure , Fibroblasts/physiology , Fibroblasts/ultrastructure , Fibrosis , Gene Expression Regulation, Developmental , Heart Ventricles , Human Umbilical Vein Endothelial Cells , Humans , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/ultrastructure , Injections , Laminin , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Morphogenesis , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Oxygen Consumption , Proteoglycans , Rats , Rats, Sprague-Dawley , Transfection , Ultrasonography
13.
J Mol Cell Cardiol ; 74: 231-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25009075

ABSTRACT

The translation of cell-based therapies for ischemic tissue repair remains limited by several factors, including poor cell survival and limited target site retention. Advances in nanotechnology enable the development of specifically designed delivery matrices to address these limitations and thereby improve the efficacy of cell-based therapies. Given the relevance of integrin signaling for cellular homeostasis, we developed an injectable, bioactive peptide-based nanofiber matrix that presents an integrin-binding epitope derived from fibronectin, and evaluated its feasibility as a supportive artificial matrix for bone marrow-derived pro-angiogenic cells (BMPACs) used as a therapy in ischemic tissue repair. Incubation of BMPACs with these peptide nanofibers in vitro significantly attenuated apoptosis while enhancing proliferation and adhesion. Pro-angiogenic function was enhanced, as cells readily formed tubes. These effects were, in part, mediated via p38, and p44/p42 MAP kinases, which are downstream pathways of focal adhesion kinase. In a murine model of hind limb ischemia, an intramuscular injection of BMPACs within this bioactive peptide nanofiber matrix resulted in greater retention of cells, enhanced capillary density, increased limb perfusion, reduced necrosis/amputation, and preserved function of the ischemic limb compared to treatment with cells alone. This self-assembling, bioactive peptide nanofiber matrix presenting an integrin-binding domain of fibronectin improves regenerative efficacy of cell-based strategies in ischemic tissue by enhancing cell survival, retention, and reparative functions.


Subject(s)
Bone Marrow Cells/cytology , Epitopes/metabolism , Fibronectins/metabolism , Ischemia/therapy , Nanofibers/administration & dosage , Peptides/administration & dosage , Animals , Biocompatible Materials , Bone Marrow Cells/metabolism , Cell Survival , Cell- and Tissue-Based Therapy/methods , Epitopes/chemistry , Fibronectins/chemistry , Gene Expression , Hindlimb/blood supply , Hindlimb/drug effects , Hindlimb/injuries , Integrins/metabolism , Ischemia/pathology , Male , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nanofibers/chemistry , Neovascularization, Physiologic , Peptides/chemical synthesis , Peptides/metabolism , Protein Binding , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Am J Physiol Heart Circ Physiol ; 306(1): H88-100, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24186100

ABSTRACT

Although the development of abnormal myocardial mechanics represents a key step during the transition from hypertension to overt heart failure (HF), the underlying ultrastructural and cellular basis of abnormal myocardial mechanics remains unclear. We therefore investigated how changes in transverse (T)-tubule organization and the resulting altered intracellular Ca(2+) cycling in large cell populations underlie the development of abnormal myocardial mechanics in a model of chronic hypertension. Hearts from spontaneously hypertensive rats (SHRs; n = 72) were studied at different ages and stages of hypertensive heart disease and early HF and were compared with age-matched control (Wistar-Kyoto) rats (n = 34). Echocardiography, including tissue Doppler and speckle-tracking analysis, was performed just before euthanization, after which T-tubule organization and Ca(2+) transients were studied using confocal microscopy. In SHRs, abnormalities in myocardial mechanics occurred early in response to hypertension, before the development of overt systolic dysfunction and HF. Reduced longitudinal, circumferential, and radial strain as well as reduced tissue Doppler early diastolic tissue velocities occurred in concert with T-tubule disorganization and impaired Ca(2+) cycling, all of which preceded the development of cardiac fibrosis. The time to peak of intracellular Ca(2+) transients was slowed due to T-tubule disruption, providing a link between declining cell ultrastructure and abnormal myocardial mechanics. In conclusion, subclinical abnormalities in myocardial mechanics occur early in response to hypertension and coincide with the development of T-tubule disorganization and impaired intracellular Ca(2+) cycling. These changes occur before the development of significant cardiac fibrosis and precede the development of overt cardiac dysfunction and HF.


Subject(s)
Heart Failure/physiopathology , Hypertension/physiopathology , Myocardium/pathology , Myocytes, Cardiac/ultrastructure , Sarcolemma/ultrastructure , Animals , Blood Pressure , Calcium/metabolism , Calcium Signaling , Fibrosis/physiopathology , Heart Failure/diagnostic imaging , Heart Failure/pathology , Heart Rate , Hypertension/diagnostic imaging , Hypertension/pathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Inbred SHR , Rats, Wistar , Ultrasonography
15.
PLoS One ; 8(11): e80809, 2013.
Article in English | MEDLINE | ID: mdl-24278323

ABSTRACT

The actin-associated protein Pdlim7 is essential for heart and fin development in zebrafish; however, the expression and function of this PDZ-LIM family member in the mammal has remained unclear. Here, we show that Pdlim7 predominantly localizes to actin-rich structures in mice including the heart, vascular smooth muscle, and platelets. To test the requirement for Pdlim7 in mammalian development and function, we analyzed a mouse strain with global genetic inactivation of Pdlim7. We demonstrate that Pdlim7 loss-of-function leads to significant postnatal mortality. Inactivation of Pdlim7 does not disrupt cardiac development, but causes mild cardiac dysfunction in adult mice. Adult Pdlim7(-/-) mice displayed increased mitral and tricuspid valve annulus to body weight ratios. These structural aberrations in Pdlim7(-/-) mice were supported by three-dimensional reconstructions of adult cardiac valves, which revealed increased surface area to volume ratios for the mitral and tricuspid valve leaflets. Unexpectedly, we found that loss of Pdlim7 triggers systemic venous and arterial thrombosis, leading to significant mortality shortly after birth in Pdlim7(+/-) (11/60) and Pdlim7(-/-) (19/35) mice. In line with a prothrombotic phenotype, adult Pdlim7(-/-) mice exhibit dramatically decreased tail bleed times compared to controls. These findings reveal a novel and unexpected function for Pdlim7 in maintaining proper hemostasis in neonatal and adult mice.


Subject(s)
Cytoskeletal Proteins/deficiency , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Heart/physiopathology , Hemostasis , Intracellular Signaling Peptides and Proteins/deficiency , LIM Domain Proteins/deficiency , Actins/metabolism , Aging/pathology , Animals , Blood Cell Count , Blood Platelets/metabolism , Blood Platelets/pathology , Crosses, Genetic , Cytoskeletal Proteins/metabolism , Embryonic Development , Female , Heart/embryology , Heart Defects, Congenital/blood , Heart Defects, Congenital/complications , Heart Valves/abnormalities , Heart Valves/pathology , Heterozygote , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Male , Mice , Thrombosis/blood , Thrombosis/complications , Thrombosis/pathology , Thrombosis/physiopathology , Weaning
16.
Am J Physiol Heart Circ Physiol ; 305(7): H1068-79, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23873796

ABSTRACT

The treatment of heart failure (HF) is challenging and morbidity and mortality are high. The goal of this study was to determine if inhibition of the late Na(+) current with ranolazine during early hypertensive heart disease might slow or stop disease progression. Spontaneously hypertensive rats (aged 7 mo) were subjected to echocardiographic study and then fed either control chow (CON) or chow containing 0.5% ranolazine (RAN) for 3 mo. Animals were then restudied, and each heart was removed for measurements of t-tubule organization and Ca(2+) transients using confocal microscopy of the intact heart. RAN halted left ventricular hypertrophy as determined from both echocardiographic and cell dimension (length but not width) measurements. RAN reduced the number of myocytes with t-tubule disruption and the proportion of myocytes with defects in intracellular Ca(2+) cycling. RAN also prevented the slowing of the rate of restitution of Ca(2+) release and the increased vulnerability to rate-induced Ca(2+) alternans. Differences between CON- and RAN-treated animals were not a result of different expression levels of voltage-dependent Ca(2+) channel 1.2, sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a, ryanodine receptor type 2, Na(+)/Ca(2+) exchanger-1, or voltage-gated Na(+) channel 1.5. Furthermore, myocytes with defective Ca(2+) transients in CON rats showed improved Ca(2+) cycling immediately upon acute exposure to RAN. Increased late Na(+) current likely plays a role in the progression of cardiac hypertrophy, a key pathological step in the development of HF. Early, chronic inhibition of this current slows both hypertrophy and development of ultrastructural and physiological defects associated with the progression to HF.


Subject(s)
Acetanilides/pharmacology , Calcium Signaling/drug effects , Hypertension/drug therapy , Myocytes, Cardiac/drug effects , Piperazines/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Sodium/metabolism , Animals , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/prevention & control , Hypertension/complications , Hypertension/diagnostic imaging , Hypertension/metabolism , Hypertension/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Male , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/drug effects , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Ranolazine , Rats , Rats, Inbred SHR , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium Channels/metabolism , Sodium-Calcium Exchanger/drug effects , Sodium-Calcium Exchanger/metabolism , Time Factors , Ultrasonography
17.
Circulation ; 127(1): 63-73, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23204107

ABSTRACT

BACKGROUND: CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. METHODS AND RESULTS: Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4(+) cells were mobilized for just 1 day in both groups, but the mobilization of sca1(+)/flk1(+) cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1(+)/flk1(+) cell mobilization without altering the mobilization of CXCR4(+) cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. CONCLUSIONS: AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Subject(s)
Heterocyclic Compounds/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines , Bone Marrow Transplantation , Cardiotonic Agents/pharmacology , Cyclams , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic/drug effects , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Nitric Oxide Synthase Type III/genetics , Recovery of Function/drug effects , Signal Transduction/drug effects
18.
Circulation ; 126(21): 2491-501, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23081990

ABSTRACT

BACKGROUND: Secretoneurin is a neuropeptide located in nerve fibers along blood vessels, is upregulated by hypoxia, and induces angiogenesis. We tested the hypothesis that secretoneurin gene therapy exerts beneficial effects in a rat model of myocardial infarction and evaluated the mechanism of action on coronary endothelial cells. METHODS AND RESULTS: In vivo secretoneurin improved left ventricular function, inhibited remodeling, and reduced scar formation. In the infarct border zone, secretoneurin induced coronary angiogenesis, as shown by increased density of capillaries and arteries. In vitro secretoneurin induced capillary tubes, stimulated proliferation, inhibited apoptosis, and activated Akt and extracellular signal-regulated kinase in coronary endothelial cells. Effects were abrogated by a vascular endothelial growth factor (VEGF) antibody, and secretoneurin stimulated VEGF receptors in these cells. Secretoneurin furthermore increased binding of VEGF to endothelial cells, and binding was blocked by heparinase, indicating that secretoneurin stimulates binding of VEGF to heparan sulfate proteoglycan binding sites. Additionally, secretoneurin increased binding of VEGF to its coreceptor neuropilin-1. In endothelial cells, secretoneurin also stimulated fibroblast growth factor receptor-3 and insulin-like growth factor-1 receptor, and in coronary vascular smooth muscle cells, we observed stimulation of VEGF receptor-1 and fibroblast growth factor receptor-3. Exposure of cardiac myocytes to hypoxia and ischemic heart after myocardial infarction revealed increased secretoneurin messenger RNA and protein. CONCLUSIONS: Our data show that secretoneurin acts as an endogenous stimulator of VEGF signaling in coronary endothelial cells by enhancing binding of VEGF to low-affinity binding sites and neuropilin-1 and stimulates further growth factor receptors like fibroblast growth factor receptor-3. Our in vivo findings indicate that secretoneurin may be a promising therapeutic tool in ischemic heart disease.


Subject(s)
Disease Models, Animal , Endothelium, Vascular/drug effects , Myocardial Infarction/drug therapy , Neovascularization, Physiologic/drug effects , Neuropeptides/administration & dosage , Secretogranin II/administration & dosage , Vascular Endothelial Growth Factor A/physiology , Animals , Coronary Vessels/drug effects , Coronary Vessels/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Genetic Therapy/methods , Humans , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/physiology , Neuropeptides/genetics , Plasmids/administration & dosage , Plasmids/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Rats , Secretogranin II/genetics , Signal Transduction/physiology
19.
Circ Res ; 111(3): 312-21, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22581926

ABSTRACT

RATIONALE: Ischemic cardiovascular disease represents one of the largest epidemics currently facing the aging population. Current literature has illustrated the efficacy of autologous, stem cell therapies as novel strategies for treating these disorders. The CD34+ hematopoetic stem cell has shown significant promise in addressing myocardial ischemia by promoting angiogenesis that helps preserve the functionality of ischemic myocardium. Unfortunately, both viability and angiogenic quality of autologous CD34+ cells decline with advanced age and diminished cardiovascular health. OBJECTIVE: To offset age- and health-related angiogenic declines in CD34+ cells, we explored whether the therapeutic efficacy of human CD34+ cells could be enhanced by augmenting their secretion of the known angiogenic factor, sonic hedgehog (Shh). METHODS AND RESULTS: When injected into the border zone of mice after acute myocardial infarction, Shh-modified CD34+ cells (CD34(Shh)) protected against ventricular dilation and cardiac functional declines associated with acute myocardial infarction. Treatment with CD34(Shh) also reduced infarct size and increased border zone capillary density compared with unmodified CD34 cells or cells transfected with the empty vector. CD34(Shh) primarily store and secrete Shh protein in exosomes and this storage process appears to be cell-type specific. In vitro analysis of exosomes derived from CD34(Shh) revealed that (1) exosomes transfer Shh protein to other cell types, and (2) exosomal transfer of functional Shh elicits induction of the canonical Shh signaling pathway in recipient cells. CONCLUSIONS: Exosome-mediated delivery of Shh to ischemic myocardium represents a major mechanism explaining the observed preservation of cardiac function in mice treated with CD34(Shh) cells.


Subject(s)
Antigens, CD34/administration & dosage , Hedgehog Proteins/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Myocardial Infarction/surgery , Animals , Antigens, CD34/therapeutic use , Cells, Cultured , Hedgehog Proteins/therapeutic use , Humans , Male , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Myocardial Infarction/physiopathology , NIH 3T3 Cells , Ventricular Dysfunction/physiopathology , Ventricular Dysfunction/surgery
20.
J Invest Dermatol ; 132(3 Pt 1): 711-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22048734

ABSTRACT

The antagonism of CXC-chemokine receptor 4 (CXCR4) with AMD3100 improves cardiac performance after myocardial infarction by augmenting the recruitment of endothelial progenitor cells (EPCs) from the bone marrow to the regenerating vasculature. We investigated whether AMD3100 may accelerate diabetes-impaired wound healing through a similar mechanism. Skin wounds were made on the backs of leptin receptor-deficient mice and treated with AMD3100 or saline. Fourteen days after treatment, wound closure was significantly more complete in AMD3100-treated mice (AMD3100: 87.0 ± 2.6%, saline: 33.1 ± 1.8%; P<0.0001) and was accompanied by greater collagen fiber formation, capillary density, smooth muscle-containing vessel density, and monocyte/macrophage infiltration. On day 7 after treatment, AMD3100 was associated with higher circulating EPC and macrophage counts, and with significantly upregulated mRNA levels of stromal cell-derived factor 1 and platelet-derived growth factor B in the wound bed. AMD3100 also promoted macrophage proliferation and phagocytosis and the migration and proliferation of diabetic mouse primary dermal fibroblasts and 3T3 fibroblasts, which express very little CXCR4. In conclusion, a single topical application of AMD3100 promoted wound healing in diabetic mice by increasing cytokine production, mobilizing bone marrow EPCs, and enhancing the activity of fibroblasts and monocytes/macrophages, thereby increasing both angiogenesis and vasculogenesis. Not all of the AMD3100-mediated effects evolved through CXCR4 antagonism.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Heterocyclic Compounds/therapeutic use , Receptors, CXCR4/antagonists & inhibitors , Skin/drug effects , Skin/injuries , Wound Healing/drug effects , Animals , Benzylamines , Capillaries/growth & development , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL12/biosynthesis , Collagen/biosynthesis , Cyclams , Female , Fibroblasts/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Phagocytosis/drug effects , Proto-Oncogene Proteins c-sis/biosynthesis , Receptors, Leptin/deficiency , Skin/pathology , Stem Cells , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...