Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Antioxid Redox Signal ; 38(4-6): 261-281, 2023 02.
Article in English | MEDLINE | ID: mdl-35950616

ABSTRACT

Aims: The objective of this study is to identify mechanisms for adipose stromal vascular fraction's (SVF) restorative effects on vasodilation in aging-induced coronary microvascular disease (CMD). We hypothesize that reactive oxygen species (ROS) diminish ß1-adrenergic receptor (ß1ADR)- and flow-mediated dilation (FMD) in coronary arterioles, reversible by SVF and adipose-derived stem cells (ADSCs). Results: SVF attenuates aging-induced chronic accumulation of ROS and pro-oxidant gene and protein expression with enhancement of antioxidant gene and protein expression and glutathione, but not nitric oxide. ADSCs attenuate hydrogen peroxide while restoring nitric oxide and glutathione. Mass spectrometry of SVF- and ADSC-conditioned media reveals abundant antioxidant proteins suggesting a paracrine mechanism. FMD and ß1ADR-mediated dilation diminished with aging, restored with SVF and ADSCs. FMD was restored by a switch in the acute signaling mediator from hydrogen peroxide in aging to peroxynitrite with SVF and ADSCs. Vasorelaxation to ß1ADR-agonism was mechanistically linked with hydrogen peroxide, nitric oxide, and glutathione. Exogenous ROS eliminates isoproterenol-mediated dilation in youth that is blocked by inhibition of pro-desensitization and internalization proteins while nitric oxide enhances isoproterenol-mediated dilation in aging. Innovation: We introduce a novel mechanism by which ROS impacts ß1ADR trafficking: the ROS/RNS-ß1ADR desensitization and internalization axis. Aging-induced ROS shunts ß1ADR from the plasma membrane into endosomes. SVF reduces oxidative burden, restoring functional ß1ADR. Conclusions: SVF (and ADSCs to a lesser extent) reduce oxidative stress, and restore flow- and ß1ADR-mediated vasodilation in aging. SVF represents a promising therapeutic strategy for CMD by addressing root cause of pathology; that is, oxidative stress-mediated hyperconstriction. Antioxid. Redox Signal. 38, 261-281.


Subject(s)
Stromal Vascular Fraction , Vasodilation , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Isoproterenol/metabolism , Isoproterenol/pharmacology , Oxidative Stress , Oxidation-Reduction , Glutathione/metabolism , Adipose Tissue/metabolism
2.
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
3.
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
5.
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
6.
Geroscience ; 44(1): 329-348, 2022 02.
Article in English | MEDLINE | ID: mdl-34608562

ABSTRACT

Our past study showed that coronary arterioles isolated from adipose-derived stromal vascular fraction (SVF)-treated rats showed amelioration of the age-related decrease in vasodilation to beta-adrenergic receptor (ß-AR) agonist and improved ß-AR-dependent coronary flow and microvascular function in a model of advanced age. We hypothesized that intravenously (i.v.) injected SVF improves coronary microvascular function in aged rats by re-establishing the equilibrium of the negative regulators of the internal adrenergic signaling cascade, G-protein receptor kinase 2 (GRK2) and G-alpha inhibitory (Gαi) proteins, back to youthful levels. Female Fischer-344 rats aged young (3 months, n = 24), old (24 months, n = 26), and old animals that received 1 × 107 green fluorescent protein (GFP+) SVF cells (O + SVF, n = 11) 4 weeks prior to sacrifice were utilized. Overnight urine was collected prior to sacrifice for catecholamine measurements. Cardiac samples were used for western blotting while coronary arterioles were isolated for pressure myography studies, immunofluorescence staining, and RNA sequencing. Coronary microvascular levels of the ß1 adrenergic receptor are decreased with advancing age, but this decreased expression was rescued by SVF treatment. Aging led to a decrease in phosphorylated GRK2 in cardiomyocytes vs. young control with restoration of phosphorylation status by SVF. In vessels, there was no change in genetic transcription (RNAseq) or protein expression (immunofluorescence); however, inhibition of GRK2 (paroxetine) led to improved vasodilation to norepinephrine in the old control (OC) and O + SVF, indicating greater GRK2 functional inhibition of ß1-AR in aging. SVF works to improve adrenergic-mediated vasodilation by restoring the ß1-AR population and mitigating signal cascade inhibitors to improve vasodilation.


Subject(s)
Aging , Cell- and Tissue-Based Therapy , Aging/pathology , Animals , Coronary Circulation , Female , G-Protein-Coupled Receptor Kinase 2/physiology , Microcirculation , Rats , Receptors, Adrenergic, beta-1/physiology , Vasodilation
7.
Antioxid Redox Signal ; 35(12): 974-1015, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34314229

ABSTRACT

Significance: The vasculature responds to the respiratory needs of tissue by modulating luminal diameter through smooth muscle constriction or relaxation. Coronary perfusion, diastolic function, and coronary flow reserve are drastically reduced with aging. This loss of blood flow contributes to and exacerbates pathological processes such as angina pectoris, atherosclerosis, and coronary artery and microvascular disease. Recent Advances: Increased attention has recently been given to defining mechanisms behind aging-mediated loss of vascular function and development of therapeutic strategies to restore youthful vascular responsiveness. The ultimate goal aims at providing new avenues for symptom management, reversal of tissue damage, and preventing or delaying of aging-induced vascular damage and dysfunction in the first place. Critical Issues: Our major objective is to describe how aging-associated mitochondrial dysfunction contributes to endothelial and smooth muscle dysfunction via dysregulated reactive oxygen species production, the clinical impact of this phenomenon, and to discuss emerging therapeutic strategies. Pathological changes in regulation of mitochondrial oxidative and nitrosative balance (Section 1) and mitochondrial dynamics of fission/fusion (Section 2) have widespread effects on the mechanisms underlying the ability of the vasculature to relax, leading to hyperconstriction with aging. We will focus on flow-mediated dilation, endothelial hyperpolarizing factors (Sections 3 and 4), and adrenergic receptors (Section 5), as outlined in Figure 1. The clinical implications of these changes on major adverse cardiac events and mortality are described (Section 6). Future Directions: We discuss antioxidative therapeutic strategies currently in development to restore mitochondrial redox homeostasis and subsequently vascular function and evaluate their potential clinical impact (Section 7). Antioxid. Redox Signal. 35, 974-1015.


Subject(s)
Aging/metabolism , Endothelium, Vascular/metabolism , Mitochondria/metabolism , Animals , Humans , Oxidation-Reduction
8.
Cardiovasc Eng Technol ; 11(5): 587-604, 2020 10.
Article in English | MEDLINE | ID: mdl-32710379

ABSTRACT

PURPOSE: The objective of this study was to reprogram human adipogenic mesenchymal stem cells (hADMSCs) to form Purkinje cells and to use the reprogrammed Purkinje cells to bioprint Purkinje networks. METHODS: hADMSCs were reprogrammed to form Purkinje cells using a multi-step process using transcription factors ETS2 and MESP1 to first form cardiac progenitor stem cells followed by SHOX2 and TBX3 to form Purkinje cells. A novel bioprinting method was developed based on Pluronic acid as the sacrificial material and type I collagen as the structural material. The reprogrammed Purkinje cells were used in conjunction with the novel bioprinting method to bioprint Purkinje networks. Printed constructs were evaluated for retention of functional protein connexin 40 (Cx40) and ability to undergo membrane potential changes in response to physiologic stimulus. RESULTS: hADMSCs were successfully reprogrammed to form Purkinje cells based on the expression pattern of IRX3, IRX5, SEMA and SCN10. Reprogrammed purkinje cells were incorporated into a collagen type-1 bioink and the left ventricular Purkinje network was printed using anatomical images of the bovine Purkinje system as reference. Optimization studies demonstrated that 1.8 mg/mL type-I collagen at a seeding density of 300,000 cells per 200 µL resulted in the most functional bioprinted Purkinje networks. Furthermore, bioprinted Purkinje networks formed continuous syncytium, retained expression of vital functional gap junction protein Cx40 post-print, and exhibited membrane potential changes in response to electric stimulation and acetylcholine evaluated by DiBAC4(5), an electrically responsive dye. CONCLUSION: Based on the results of this study, hADMSCs were successfully reprogrammed to form Purkinje cells and bioprinted to form Purkinje networks.


Subject(s)
Adipogenesis , Bioprinting , Cellular Reprogramming Techniques , Cellular Reprogramming , Mesenchymal Stem Cells/physiology , Printing, Three-Dimensional , Purkinje Fibers/physiology , Cell Communication , Cells, Cultured , Humans , Phenotype , Purkinje Fibers/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
9.
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
10.
Aging (Albany NY) ; 11(13): 4561-4578, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31296794

ABSTRACT

Our past study showed that a single tail vein injection of adipose-derived stromal vascular fraction (SVF) into old rats was associated with improved dobutamine-mediated coronary flow reserve. We hypothesize that i.v. injection of SVF improves coronary microvascular function in aged rats via alterations in beta adrenergic microvascular signaling. Female Fischer-344 rats aged young (3 months, n=32) and old (24 months, n=30) were utilized, along with two cell therapies intravenously injected in old rats four weeks prior to sacrifice: 1x107 green fluorescent protein (GFP+) SVF cells (O+SVF, n=21), and 5x106 GFP+ bone-marrow mesenchymal stromal cells (O+BM, n=6), both harvested from young donors. Cardiac ultrasound and pressure-volume measurements were obtained, and coronary arterioles were isolated from each group for microvessel reactivity studies and immunofluorescence staining. Coronary flow reserve decreased with advancing age, but this effect was rescued by the SVF treatment in the O+SVF group. Echocardiography showed an age-related diastolic dysfunction that was improved with SVF to a greater extent than with BM treatment. Coronary arterioles isolated from SVF-treated rats showed amelioration of the age-related decrease in vasodilation to a non-selective ß-AR agonist. I.v. injected SVF cells improved ß-adrenergic receptor-dependent coronary flow and microvascular function in a model of advanced age.


Subject(s)
Adipose Tissue/cytology , Age Factors , Arterioles/cytology , Receptors, Adrenergic, beta-1/metabolism , Stromal Cells/cytology , Animals , Female , Fractional Flow Reserve, Myocardial , Green Fluorescent Proteins , Injections, Intravenous , Luminescent Agents , Mesenchymal Stem Cells/cytology , Rats , Rats, Inbred F344 , Signal Transduction , Vasodilation
11.
Echocardiography ; 35(1): 90-99, 2018 01.
Article in English | MEDLINE | ID: mdl-29082558

ABSTRACT

BACKGROUND: Quantification of defect size and shunt flow is an important aspect of ventricular septal defect (VSD) evaluation. This study compared three-dimensional echocardiography (3DE) with the current clinical standard two-dimensional echocardiography (2DE) for quantifying defect area and tested the feasibility of real time 3D color Doppler echocardiography (RT3D-CDE) for quantifying shunt volume of irregular shaped and multiple VSDs. METHODS: Latex balloons were sutured into the ventricles of 32 freshly harvested porcine hearts and were connected with tubing placed in septal perforations. Tubing was varied in area (0.13-5.22 cm²), number (1-3), and shape (circle, oval, crescent, triangle). A pulsatile pump was used to pump "blood" through the VSD (LV to RV) at stroke volumes of 30-70 mL with a stroke rate of 60 bpm. Two-dimensional echocardiography (2DE), 3DE, and RT3D-CDE images were acquired from the right side of the phantom. RESULTS: For circular VSDs, both 2DE and 3DE area measurements were consistent with the actual areas (R² = 0.98 vs 0.99). For noncircular/multiple VSDs, 3DE correlated with the actual area more closely than 2DE (R² = 0.99 vs 0.44). Shunt volumes obtained using RT3D-CDE positively correlated with pumped stroke volumes (R² = 0.96). CONCLUSIONS: Three-dimensional echocardiography (3DE) is a feasible method for determining VSD area and is more accurate than 2DE for evaluating the area of multiple or noncircular VSDs. Real-time 3D color Doppler echocardiography (RT3D-CDE) is a feasible method for quantifying the shunt volume of multiple or noncircular VSDs.


Subject(s)
Echocardiography/methods , Heart Septal Defects, Ventricular/diagnostic imaging , Heart Ventricles/diagnostic imaging , Animals , Disease Models, Animal , Echocardiography, Doppler, Color/methods , Echocardiography, Three-Dimensional/methods , Feasibility Studies , Heart Septal Defects, Ventricular/pathology , Heart Ventricles/pathology , In Vitro Techniques , Reproducibility of Results , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...