Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
J Surg Res ; 295: 28-40, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37979234

ABSTRACT

INTRODUCTION: Graft loss in vascularized composite allotransplantation (VCA) is more often associated with vasculopathy and chronic rejection (CR) than acute cellular rejection (ACR). We present a rat osteomyocutaneous flap model using titrated tacrolimus administration that mimics the graft rejection patterns in our clinical hand transplant program. Comparison of outcomes in these models support a role for ischemia reperfusion injury (IRI) and microvascular changes in CR of skin and large-vessel vasculopathy. The potential of the surgical models for investigating mechanisms of rejection and vasculopathy in VCA and treatment interventions is presented. MATERIALS AND METHODS: Four rodent groups were evaluated: syngeneic controls (Group 1), allogeneic transient immunosuppression (Group 2), allogeneic suboptimal immunosuppression (Group 3), and allogeneic standard immunosuppression (Group 4). Animals were monitored for ACR, vasculopathy, and CR of the skin. RESULTS: Transient immunosuppression resulted in severe ACR within 2 wk of tacrolimus discontinuation. Standard immunosuppression resulted in minimal rejection but subclinical microvascular changes, including capillary thrombosis and luminal narrowing in arterioles in the donor skin. Further reduction in tacrolimus dose led to femoral vasculopathy and CR of the skin. Surprisingly, femoral vasculopathy was also observed in the syngeneic control group. CONCLUSIONS: Titration of tacrolimus in the allogeneic VCA model resulted in presentations of rejection and vasculopathy similar to those in patients and suggests vasculopathy starts at the microvascular level. This adjustable experimental model will allow the study of variables and interventions, such as external trauma or complement blockade, that may initiate or mitigate vasculopathy and CR in VCA.


Subject(s)
Tacrolimus , Vascularized Composite Allotransplantation , Humans , Rats , Animals , Vascularized Composite Allotransplantation/adverse effects , Vascularized Composite Allotransplantation/methods , Surgical Flaps , Immunosuppression Therapy , Immune Tolerance , Graft Rejection/diagnosis , Graft Rejection/etiology , Graft Survival
2.
Am J Physiol Heart Circ Physiol ; 323(4): H749-H762, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36018760

ABSTRACT

Aging is associated with blunted coronary microvascular vasodilatory function. Previously, systemically administered adipose stromal vascular fraction (SVF) therapy reversed aging-induced attenuation of ß1-adrenergic- and flow-mediated dilation dependent on reducing mitochondrial reactive oxygen species. We hypothesized that SVF-mediated recovery of microvascular dilatory function is dependent on recovery of mitochondrial function, specifically by reducing mitochondrial hyperfission. Female Fischer-344 rats were allocated into young control, old control, and old + SVF therapy groups. Pressure myography, immunofluorescent staining, Western blot analysis, and RNA sequencing were performed to determine coronary microvascular mitochondrial dynamics and function. Gene and protein expression of fission-mediator DRP-1 was enhanced with aging but reversed by SVF therapy. SVF facilitated an increase in fusion-mediator MFN-1 gene and protein expression. Mitochondrial morphology was characterized as rod-like and densely networked in young controls, isolated circular and punctate with aging, and less circularity with partially restored mitochondrial branch density with SVF therapy. Decreased mitochondrial membrane potential and ATP bioavailability in aged animals at baseline and during flow-mediated dilation were reversed by SVF and accompanied with enhanced oxygen consumption. Dilation to norepinephrine and flow in young controls were dependent on uninhibited mitochondrial fusion, whereas inhibiting fission did not restore aged microvessel response to norepinephrine or flow. SVF-mediated recovery of ß-adrenergic function was dependent on uninhibited mitochondrial fusion, whereas recovery of flow-mediated dilation was dependent on maintained mitochondrial fission. Impaired dilation in aging is mitigated by SVF therapy, which recovers mitochondrial function and fission/fusion balance.NEW & NOTEWORTHY We elucidated the consequences of aging on coronary microvascular mitochondrial health as well as SVF's ability to reverse these effects. Aging shifts gene/protein expression and mitochondrial morphology indicating hyperfission, alongside attenuated mitochondrial membrane potential and ATP bioavailability, all reversed using SVF therapy. Mitochondrial membrane potential and ATP levels correlated with vasodilatory efficiency. Mitochondrial dysfunction is a contributing pathological factor in aging that can be targeted by therapeutic SVF to preserve microvascular dilative function.


Subject(s)
Adipose Tissue , Stromal Cells , Adenosine Triphosphate/metabolism , Adipose Tissue/metabolism , Adrenergic Agents , Animals , Female , Mitochondria , Norepinephrine/metabolism , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Stromal Cells/metabolism , Stromal Vascular Fraction
3.
Am J Physiol Heart Circ Physiol ; 323(6): H1167-H1175, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36306213

ABSTRACT

Microvascular disease plays a critical role in systemic end-organ dysfunction, and treatment of microvascular pathologies may greatly reduce cardiovascular morbidity and mortality. The Call for Papers collection: New Developments in Translational Microcirculatory Research highlights key advances in our understanding of the role of microvessels in the development of chronic diseases as well as therapeutic strategies to enhance microvascular function. This Mini Review provides a concise summary of these advances and draws from other relevant research to provide the most up-to-date information on the influence of cutaneous, cerebrovascular, coronary, and peripheral microcirculation on the pathophysiology of obesity, hypertension, cardiovascular aging, peripheral artery disease, and cognitive impairment. In addition to these disease- and location-dependent research articles, this Call for Papers includes state-of-the-art reviews on coronary endothelial function and assessment of microvascular health in different organ systems, with an additional focus on establishing rigor and new advances in clinical trial design. These articles, combined with original research evaluating cellular, exosomal, pharmaceutical, exercise, heat, and dietary interventional therapies, establish the groundwork for translating microcirculatory research from bench to bedside. Although numerous studies in this collection are focused on human microcirculation, most used robust preclinical models to probe mechanisms of pathophysiology and interventional benefits. Future work focused on translating these findings to humans are necessary for finding clinical strategies to prevent and treat microvascular dysfunction.


Subject(s)
Hypertension , Peripheral Vascular Diseases , Humans , Microcirculation/physiology , Microvessels , Endothelium
4.
Am J Physiol Heart Circ Physiol ; 322(4): H647-H680, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35179976

ABSTRACT

Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.


Subject(s)
Microvessels , Vascular Diseases , Animals , Endothelium, Vascular/metabolism , Oxidative Stress , Regeneration , Vascular Diseases/metabolism
5.
Microcirculation ; 29(6-7): e12758, 2022 10.
Article in English | MEDLINE | ID: mdl-35466504

ABSTRACT

OBJECTIVE: The objective of the study is to demonstrate the innovation and utility of mesenteric tissue culture for discovering the microvascular growth dynamics associated with adipose-derived stromal vascular fraction (SVF) transplantation. Understanding how SVF cells contribute to de novo vessel growth (i.e., neovascularization) and host network angiogenesis motivates the need to make observations at single-cell and network levels within a tissue. METHODS: Stromal vascular fraction was isolated from the inguinal adipose of adult male Wistar rats, labeled with DiI, and seeded onto adult Wistar rat mesentery tissues. Tissues were then cultured in MEM + 10% FBS for 3 days and labeled for BSI-lectin to identify vessels. Alternatively, SVF and tissues from green fluorescent-positive (GFP) Sprague Dawley rats were used to track SVF derived versus host vasculature. RESULTS: Stromal vascular fraction-treated tissues displayed a dramatically increased vascularized area compared to untreated tissues. DiI and GFP+ tracking of SVF identified neovascularization involving initial segment formation, radial outgrowth from central hub-like structures, and connection of segments. Neovascularization was also supported by the formation of segments in previously avascular areas. New segments characteristic of SVF neovessels contained endothelial cells and pericytes. Additionally, a subset of SVF cells displayed the ability to associate with host vessels and the presence of SVF increased host network angiogenesis. CONCLUSIONS: The results showcase the use of the rat mesentery culture model as a novel tool for elucidating SVF cell transplant dynamics and highlight the impact of model selection for visualization.


Subject(s)
Endothelial Cells , Stromal Cells , Rats , Male , Animals , Stromal Vascular Fraction , Rats, Sprague-Dawley , Rats, Wistar , Microvessels , Adipose Tissue/blood supply , Neovascularization, Pathologic , Mesentery
6.
J Vasc Res ; 59(6): 343-357, 2022.
Article in English | MEDLINE | ID: mdl-36075199

ABSTRACT

Adipose-derived stromal vascular fraction (SVF) has emerged as a potential regenerative therapy, but few studies utilize SVF in a setting of advanced age. Additionally, the specific cell population in SVF providing therapeutic benefit is unknown. We hypothesized that aging would alter the composition of cell populations present in SVF and its ability to promote angiogenesis following injury, a mechanism that is T cell-mediated. SVF isolated from young and old Fischer 344 rats was examined with flow cytometry for cell composition. Mesenteric windows from old rats were isolated following exteriorization-induced (EI) hypoxic injury and intravenous injection of one of four cell therapies: (1) SVF from young or (2) old donors, (3) SVF from old donors depleted of or (4) enriched for T cells. Advancing age increased the SVF T-cell population but reduced revascularization following injury. Both young and aged SVF incorporated throughout the host mesenteric microvessels, but only young SVF significantly increased vascular area following EI. This study highlights the effect of donor age on SVF angiogenic efficacy and demonstrates how the ex vivo mesenteric-window model can be used in conjunction with SVF therapy to investigate its contribution to angiogenesis.


Subject(s)
Adipose Tissue , Stromal Cells , Rats , Animals , Stromal Vascular Fraction , Rats, Inbred F344 , Microvessels
7.
Cardiovasc Drugs Ther ; 36(3): 425-436, 2022 06.
Article in English | MEDLINE | ID: mdl-35157168

ABSTRACT

PURPOSE: Nrf2 is a nuclear transcription factor and plays an important role in the regulation of oxidative stress and inflammation. We recently demonstrated that sulforaphane (SFN) protected mice from developing pulmonary arterial hypertension (PAH) and right ventricular (RV) dysfunction by elevating cardiac Nrf2 expression and function. Here we further investigate Nrf2 dependence for SFN-mediated prevention of PAH and RV dysfunction in an Nrf2 knockout mouse model. METHODS: We used male global Nrf2-knockout mice and male C57/6 J wild type mice in the following groups: Control group received room air and vehicle control; SuHx group received SU5416 and 10% hypoxia for 4 weeks to induce PAH; SuHx+SFN group received both SuHx and sulforaphane, a Nrf2 activator, for 4 weeks. Transthoracic echocardiography was performed to quantify RV function and estimate pulmonary vascular resistance over 4 weeks. PAH was confirmed using invasive RV systolic pressure measurement at 4 weeks. RESULTS: All Nrf2 knockout mice survived the 4-week SuHx induction of PAH. SuHx caused progressive RV diastolic/systolic dysfunction and increased RV systolic pressure. The development of RV diastolic dysfunction occurred earlier in the Nrf2 knockout PAH mice when compared with the wide type PAH mice. SFN partially or completely reversed SuHx-induced RV diastolic/systolic dysfunction and increased RV systolic pressure in wild-type mice, but not in Nrf2 knockout mice. CONCLUSION: Our findings demonstrated the essential role of Nrf2 in SFN-mediated prevention of RV dysfunction and PAH, and increasing Nrf2 activity in patients with PAH may have therapeutic potential.


Subject(s)
Hypertension, Pulmonary , NF-E2-Related Factor 2 , Pulmonary Arterial Hypertension , Ventricular Dysfunction, Right , Animals , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/prevention & control , Isothiocyanates , Male , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Pulmonary Artery , Sulfoxides , Ventricular Dysfunction, Right/drug therapy , Ventricular Dysfunction, Right/prevention & control
8.
Am J Physiol Cell Physiol ; 319(1): C166-C182, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32432929

ABSTRACT

This review aims to highlight the normal physiological remodeling that occurs in healthy aging hearts, including changes that occur in contractility, conduction, valve function, large and small coronary vessels, and the extracellular matrix. These "normal" age-related changes serve as the foundation that supports decreased plasticity and limited ability for tissue remodeling during pathophysiological states such as myocardial ischemia and heart failure. This review will identify populations at greater risk for poor tissue remodeling in advanced age along with present and future therapeutic strategies that may ameliorate dysfunctional tissue remodeling in aging hearts.


Subject(s)
Healthy Aging/pathology , Heart Diseases/pathology , Myocardium/pathology , Ventricular Remodeling/physiology , Aging/metabolism , Aging/pathology , Animals , Coronary Vessels/metabolism , Coronary Vessels/pathology , Healthy Aging/metabolism , Heart Diseases/metabolism , Humans , Myocardium/metabolism
9.
Immunol Cell Biol ; 98(8): 650-666, 2020 09.
Article in English | MEDLINE | ID: mdl-32392367

ABSTRACT

The metabolically dynamic nature of healthy adipose places this tissue under regular inflammatory stress. A network of adipose-resident anti-inflammatory immune cells modulates and resolves this endogenous inflammation. Previous work in our laboratory identified a CD11b+ Gr1+ subset of these immunosuppressive adipose stromal cells in healthy mice. Myeloid-derived suppressor cells (MDSCs), typically associated with cancer and chronic inflammation, have a similar surface marker phenotype and accumulate in adipose of high-fat diet-fed mice. Given the routine inflammatory stresses on healthy adipose and the suppressive nature of the tissue-resident immune cells, we hypothesized that these CD11b+ Gr1+ cells were a genuine population of MDSCs involved in regulating tissue homeostasis. Flow cytometric analysis of these cells found that they were CD11b+ CD301- Ly6C+ Ly6G+/- and did not express traditional macrophage markers. Moreover, in vitro functional assays demonstrated that these cells suppressed αCD3/αCD28-induced T-cell proliferation, solidifying their identity as bona fide adipose-resident MDSCs. Systemic MDSC depletion altered adipose immune cell dynamics in otherwise healthy mice, increasing the number of CD4+ effector memory T cells and modifying the surface markers expressed by adipose-resident macrophages. In addition, transcription of various immunomodulatory cytokines was clearly dysregulated in the adipose of MDSC-depleted animals compared with controls. Altogether, our findings indicate that there is a population of bona fide MDSCs in the adipose of otherwise healthy mice that actively contribute to the health and immune homeostasis of this tissue.


Subject(s)
Adipose Tissue/immunology , Homeostasis/immunology , Myeloid-Derived Suppressor Cells , Animals , CD11b Antigen , Cytokines , Lymphocyte Activation , Macrophages , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes
15.
Am J Physiol Cell Physiol ; 313(2): C197-C206, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28515088

ABSTRACT

Dopamine decreases Na-K-ATPase (NKA) activity by PKC-dependent phosphorylation and endocytosis of the NKA α1. Dopamine-mediated regulation of NKA is impaired in aging and some forms of hypertension. Using opossum (OK) proximal tubule cells (PTCs), we demonstrated that sodium-hydrogen exchanger regulatory factor-1 (NHERF-1) associates with NKA α1 and dopamine-1 receptor (D1R). This association is required for the dopamine-mediated regulation of NKA. In OK cells, dopamine decreases NHERF-1 association with NKA α1 but increases its association with D1R. However, it is not known whether NHERF-1 plays a role in dopamine-mediated NKA regulation in animal models of hypertension. We hypothesized that defective dopamine-mediated regulation of NKA results from the decrease in NHERF-1 expression in rat renal PTCs isolated from animal models of hypertension [spontaneously hypertensive rats (SHRs) and aged F344 rats]. To test this hypothesis, we isolated and cultured renal PTCs from 22-mo-old F344 rats and their controls, normotensive 4-mo-old F344 rats, and SHRs and their controls, normotensive Wistar-Kyoto (WKY) rats. The results demonstrate that in both hypertensive models (SHR and aged F344), NHERF-1 expression, dopamine-mediated phosphorylation of NKA, and ouabain-inhibitable K+ transport are reduced. Transfection of NHERF-1 into PTCs from aged F344 and SHRs restored dopamine-mediated inhibition of NKA. These results suggest that decreased renal NHERF-1 expression contributes to the impaired dopamine-mediated inhibition of NKA in PTCs from animal models of hypertension.


Subject(s)
Hypertension/genetics , Kidney Tubules, Proximal/metabolism , Phosphoproteins/biosynthesis , Sodium-Hydrogen Exchangers/biosynthesis , Sodium-Potassium-Exchanging ATPase/biosynthesis , Animals , Blood Pressure/genetics , Cell Line , Disease Models, Animal , Dopamine/metabolism , Gene Expression Regulation/genetics , Humans , Hypertension/metabolism , Hypertension/pathology , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/pathology , Male , Phosphoproteins/genetics , Rats , Rats, Inbred SHR , Signal Transduction/genetics , Sodium-Hydrogen Exchangers/genetics , Sodium-Potassium-Exchanging ATPase/genetics
16.
Am J Physiol Heart Circ Physiol ; 310(11): H1842-50, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27199114

ABSTRACT

Aging and cardiovascular disease are associated with the loss of nitric oxide (NO) signaling and a decline in the ability to increase coronary blood flow reserve (CFR). Thrombospondin-1 (Thbs-1), through binding of CD47, has been shown to limit NO-dependent vasodilation in peripheral vascular beds via formation of superoxide (O2 (-)). The present study tests the hypothesis that, similar to the peripheral vasculature, blocking CD47 will improve NO-mediated vasoreactivity in coronary arterioles from aged individuals, resulting in improved CFR. Isolated coronary arterioles from young (4 mo) or old (24 mo) female Fischer 344 rats were challenged with the NO donor, DEA-NONO-ate (1 × 10(-7) to 1 × 10(-4) M), and vessel relaxation and O2 (-) production was measured before and after Thbs-1, αCD47, and/or Tempol and catalase exposure. In vivo CFR was determined in anesthetized rats (1-3% isoflurane-balance O2) via injected microspheres following control IgG or αCD47 treatment (45 min). Isolated coronary arterioles from young and old rats relax similarly to exogenous NO, but addition of 2.2 nM Thbs-1 inhibited NO-mediated vasodilation by 24% in old rats, whereas young vessels were unaffected. Thbs-1 increased O2 (-) production in coronary arterioles from rats of both ages, but this was exaggerated in old rats. The addition of CD47 blocking antibody completely restored NO-dependent vasodilation in isolated arterioles from aged rats and attenuated O2 (-) production. Furthermore, αCD47 treatment increased CFR from 9.6 ± 9.3 (IgG) to 84.0 ± 23% in the left ventricle in intact, aged animals. These findings suggest that the influence of Thbs-1 and CD47 on coronary perfusion increases with aging and may be therapeutically targeted to reverse coronary microvascular dysfunction.


Subject(s)
Aging/physiology , Arterioles/drug effects , Coronary Vessels/drug effects , Hydrazines/pharmacology , Nitric Oxide Donors/pharmacology , Thrombospondin 1/pharmacology , Vasodilation/drug effects , Animals , Endothelium, Vascular/drug effects , Female , Rats , Rats, Inbred F344
19.
Microcirculation ; 23(2): 157-67, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26661273

ABSTRACT

Advancing age will affect every individual and its impact on health deserves significant attention particularly as we address therapeutic possibilities to pathological conditions. The changes that occur in the coronary vasculature as a result of aging-related senescence set the stage upon which CVD and ischemia can escalate. Because of its importance in health, the consequences of aging on vasculature adaptation must be considered as we identify molecular targets and cell therapies for older patients. To understand the complex relationships between the coronary vasculature and the myocardium, it is important to characterize the unique aged cardiac environment in both locales independent of overlying disease. Therefore, the overall theme of this review is to highlight the biology of aging coronary vasculature and how this promotes a decreased plasticity, exacerbating insults such as ischemia. We will identify potential age-related mechanisms that may contribute to this overall loss of adaptation and regeneration and review potential therapeutic strategies to ameliorate this dysfunction.


Subject(s)
Aging/metabolism , Coronary Circulation , Microcirculation , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Aging/pathology , Animals , Humans , Myocardial Ischemia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL