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










Publication year range
1.
Transl Res ; 269: 14-30, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38453052

ABSTRACT

The progression of chronic kidney disease (CKD) often involves renal interstitial fibrosis (RIF) and subsequent loss of peritubular capillaries (PTCs), which enhances disease severity. Despite advancements in our understanding of fibrosis, effective interventions for reversing capillary loss remain elusive. Notably, RIF exhibits reduced capillary density, whereas renal cell carcinoma (RCC) shows robust angiogenesis under hypoxic conditions. Using RNA sequencing and bioinformatics, we identified differentially expressed genes (DEGs) in hypoxic human renal tubular epithelial cells (HK-2) and renal cancer cells (786-0). Analysis of altered Ras and PI3K/Akt pathways coupled with hub gene investigation revealed RAS protein activator-like 2 (RASAL2) as a key candidate. Subsequent in vitro and in vivo studies confirmed RASAL2's early-stage response in RIF, which reduced with fibrosis progression. RASAL2 suppression in HK-2 cells enhanced angiogenesis, as evidenced by increased proliferation, migration, and branching of human umbilical vein endothelial cells (HUVECs) co-cultured with HK-2 cells. In mice, RASAL2 knockdown improved Vascular endothelial growth factor A (VEGFA) and Proliferating cell nuclear antigen (PCNA) levels in unilateral ureteral occlusion (UUO)-induced fibrosis (compared to wild type). Hypoxia-inducible factor 1 alpha (HIF-1α) emerged as a pivotal mediator, substantiated by chromatin immunoprecipitation (ChIP) sequencing, with its induction linked to activation. Hypoxia increased the production of RASAL2-enriched extracellular vesicles (EVs) derived from tubular cells, which were internalized by endothelial cells, contributing to the exacerbation of PTC loss. These findings underscore RASAL2's role in mediating reduced angiogenesis in RIF and reveal a novel EV-mediated communication between hypoxic tubular- and endothelial cells, demonstrating a complex interplay between angiogenesis and fibrosis in CKD pathogenesis.


Subject(s)
Fibrosis , Humans , Animals , Mice , Male , Human Umbilical Vein Endothelial Cells/metabolism , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/pathology , Microvascular Rarefaction/genetics , Mice, Inbred C57BL , Kidney/blood supply , Kidney/pathology , Kidney/metabolism , Hypoxia/pathology , Hypoxia/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/genetics , Cell Hypoxia , Kidney Tubules/pathology , Kidney Tubules/metabolism , Cell Line , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics
2.
J Am Heart Assoc ; 13(3): e029427, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38293915

ABSTRACT

BACKGROUND: The right ventricle (RV) is at risk in patients with complex congenital heart disease involving right-sided obstructive lesions. We have shown that capillary rarefaction occurs early in the pressure-loaded RV. Here we test the hypothesis that microRNA (miR)-34a, which is induced in RV hypertrophy and RV failure (RVF), blocks the hypoxia-inducible factor-1α-vascular endothelial growth factor (VEGF) axis, leading to the attenuated angiogenic response and increased susceptibility to RV failure. METHODS AND RESULTS: Mice underwent pulmonary artery banding to induce RV hypertrophy and RVF. Capillary rarefaction occurred immediately. Although hypoxia-inducible factor-1α expression increased (0.12±0.01 versus 0.22±0.03, P=0.05), VEGF expression decreased (0.61±0.03 versus 0.22±0.05, P=0.01). miR-34a expression was most upregulated in fibroblasts (4-fold), but also in cardiomyocytes and endothelial cells (2-fold). Overexpression of miR-34a in endothelial cells increased cell senescence (10±3% versus 22±2%, P<0.05) by suppressing sirtulin 1 expression, and decreased tube formation by 50% via suppression of hypoxia-inducible factor-1α, VEGF A, VEGF B, and VEGF receptor 2. miR-34a was induced by stretch, transforming growth factor-ß1, adrenergic stimulation, and hypoxia in cardiac fibroblasts and cardiomyocytes. In mice with RVF, locked nucleic acid-antimiR-34a improved RV shortening fraction and survival half-time and restored capillarity and VEGF expression. In children with congenital heart disease-related RVF, RV capillarity was decreased and miR-34a increased 5-fold. CONCLUSIONS: In summary, miR-34a from fibroblasts, cardiomyocytes, and endothelial cells mediates capillary rarefaction by suppressing the hypoxia-inducible factor-1α-VEGF axis in RV hypertrophy/RVF, raising the potential for anti-miR-34a therapeutics in patients with at-risk RVs.


Subject(s)
Heart Defects, Congenital , Heart Failure , MicroRNAs , Microvascular Rarefaction , Child , Humans , Mice , Animals , Vascular Endothelial Growth Factor A/metabolism , Endothelial Cells/metabolism , Angiogenesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Microvascular Rarefaction/metabolism , Heart Failure/metabolism , Hypertrophy, Right Ventricular , Myocytes, Cardiac/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Heart Defects, Congenital/metabolism
3.
Am J Physiol Cell Physiol ; 326(3): C712-C723, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38223932

ABSTRACT

Fibrosis, the morphologic end-result of a plethora of chronic conditions and the scorch for organ function, has been thoroughly investigated. One aspect of its development and progression, namely the permissive role of vascular endothelium, has been overshadowed by studies into (myo)fibroblasts and TGF-ß; thus, it is the subject of the present review. It has been established that tensile forces of the extracellular matrix acting on cells are a prerequisite for mechanochemical coupling, leading to liberation of TGF-ß and formation of myofibroblasts. Increased tensile forces are prompted by elevated vascular permeability in response to diverse stressors, resulting in the exudation of fibronectin, fibrinogen/fibrin, and other proteins, all stiffening the extracellular matrix. These processes lead to the development of endothelial cells dysfunction, endothelial-to-mesenchymal transition, premature senescence of endothelial cells, perturbation of blood flow, and gradual obliteration of microvasculature, leaving behind "string" vessels. The resulting microvascular rarefaction is not only a constant companion of fibrosis but also an adjunct mechanism of its progression. The deepening knowledge of the above chain of pathogenetic events involving endothelial cells, namely increased permeability-stiffening of the matrix-endothelial dysfunction-microvascular rarefaction-tissue fibrosis, may provide a roadmap for therapeutic interventions deemed to curtail and reverse fibrosis.


Subject(s)
Endothelium, Vascular , Microvascular Rarefaction , Humans , Endothelium, Vascular/metabolism , Endothelial Cells/metabolism , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/pathology , Kidney/metabolism , Fibrosis , Transforming Growth Factor beta/metabolism
4.
Physiol Rep ; 11(6): e15643, 2023 03.
Article in English | MEDLINE | ID: mdl-36946064

ABSTRACT

Endothelial cell-selective adhesion molecule (ESAM) regulates inflammatory cell adhesion and transmigration and promotes angiogenesis. Here, we examined the role of ESAM in cardiac vascularization, inflammatory cell infiltration, and left ventricle (LV) diastolic function under basal and hemodynamic stress conditions. We employed mice with homozygous genetic deletion of ESAM (ESAM-/- ) and also performed uninephrectomy and aldosterone infusion (UNX-Aldo) to induce volume and pressure overload. Using echocardiography, we found that ESAM-/- mice display no change in systolic function. However, they develop LV diastolic dysfunction, as indicated by a significantly reduced E/A ratio (E = early, A = late mitral inflow peak velocities), increased E/e' ratio, isovolumic relaxation time (IVRT), and E wave deceleration time. An unbiased automated tracing and 3D reconstruction of coronary vasculature revealed that ESAM-/- mice had reduced coronary vascular density. Arteries of ESAM-/- mice exhibited impaired endothelial sprouting and in cultured endothelial cells siRNA-mediated ESAM knockdown reduced tube formation. Changes in ESAM-/- mice were accompanied by elevated myocardial inflammatory cytokine and myeloperoxidase-positive neutrophil levels. Furthermore, UNX-Aldo procedure in wild type mice induced LV diastolic dysfunction, which was accompanied by significantly increased serum ESAM levels. When compared to wild types, ESAM-/- mice with UNX-Aldo displayed worsening of LV diastolic function, as indicated by increased IVRT and pulmonary edema. Thus, we propose that ESAM plays a mechanistic role in proper myocardial vascularization and the maintenance of LV diastolic function under basal and hemodynamic stress conditions.


Subject(s)
Microvascular Rarefaction , Ventricular Dysfunction, Left , Mice , Animals , Endothelial Cells/metabolism , Heart Ventricles , Microvascular Rarefaction/metabolism , Heart , Ventricular Function, Left , Diastole
5.
Kidney Int ; 102(4): 780-797, 2022 10.
Article in English | MEDLINE | ID: mdl-35934136

ABSTRACT

Plasma levels of angiopoietin-2 are increased in patients with chronic kidney disease (CKD). Moreover, mouse models of progressive kidney disease also demonstrate increased angiopoietin-2 in both plasmas and kidneys. The role of dysregulated angiopoietins in the progression of kidney disease has not been thoroughly investigated. Here, we found in a cohort of 319 patients with CKD that plasma angiopoietin-2 and angiopoietin-2/angiopoietin-1 ratios were positively associated with the development of kidney failure. In mice with progressive kidney disease induced by either ureteral obstruction or ischemia-reperfusion injury, overexpression of human angiopoietin-1 in the kidney tubules not only reduced macrophage infiltration in the initial stage post-injury but also attenuated endothelial cell apoptosis, microvascular rarefaction, and fibrosis in the advanced disease stage. Notably, angiopoietin-1 attenuated chemokine C-C motif ligand 2 (CCL2) expression in the endothelial cells of the fibrosing kidneys, and these protective effects led to attenuation of functional impairment. Mechanistically, angiopoietin-1 reduced CCL2-activated macrophage migration and protected endothelial cells against cell apoptosis induced by angiopoietin-2 and Wnt ligands. Based on this, we applied L1-10, an angiopoietin-2 inhibitor, to the mouse models of progressive kidney disease and found inhibitory effects on macrophage infiltration, microvascular rarefaction, and fibrosis. Thus, we defined the detrimental impact of increased angiopoietin-2 on kidney survival of patients with CKD which appears highlighted by angiopoietin-2 induced endothelial CCL2-activated macrophage infiltration and endothelial cell apoptosis in their kidneys undergoing fibrosis.


Subject(s)
Microvascular Rarefaction , Renal Insufficiency, Chronic , Angiopoietin-1 , Angiopoietin-2/metabolism , Animals , Apoptosis , Chemokine CCL2/metabolism , Chemokines/metabolism , Endothelial Cells/pathology , Fibrosis , Humans , Kidney/pathology , Ligands , Mice , Mice, Inbred C57BL , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/pathology , Renal Insufficiency, Chronic/pathology
6.
Sci Adv ; 7(5)2021 01.
Article in English | MEDLINE | ID: mdl-33571112

ABSTRACT

Endothelial cell injury leading to microvascular rarefaction is a characteristic feature of chronic kidney disease (CKD). However, the mechanism underlying endothelial cell dropout is poorly defined. Here, we show a central role of the extracellular microenvironment in controlling endothelial cell survival and proliferation in CKD. When cultured on a decellularized kidney tissue scaffold (KTS) from fibrotic kidney, endothelial cells increased the expression of proapoptotic proteins. Proteomics profiling identified fibrillin-1 (FBN1) as a key component of the fibrotic KTS, which was up-regulated in animal models and patients with CKD. FBN1 induced apoptosis of endothelial cells and inhibited their proliferation in vitro. RNA sequencing uncovered activated integrin αvß6/transforming growth factor-ß signaling, and blocking this pathway abolished FBN1-triggered endothelial injury. In a mouse model of CKD, depletion of FBN1 ameliorated renal fibrotic lesions and mitigated vascular rarefaction. These studies illustrate that FBN1 plays a role in mediating vascular rarefaction by orchestrating a hostile microenvironment for endothelial cells.


Subject(s)
Endothelial Cells , Fibrillin-1 , Microvascular Rarefaction , Renal Insufficiency, Chronic , Animals , Cellular Microenvironment/genetics , Cellular Microenvironment/physiology , Endothelial Cells/metabolism , Female , Fibrillin-1/genetics , Fibrillin-1/metabolism , Fibrosis , Humans , Kidney/pathology , Male , Mice , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/pathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology
7.
Steroids ; 156: 108573, 2020 04.
Article in English | MEDLINE | ID: mdl-31904375

ABSTRACT

This work investigated the mechanisms induced by exercise training that may contribute to attenuate dexamethasone (DEX)-induced microvascular rarefaction and hypertension. Wistar rats underwent training protocol or were kept sedentary for 8 weeks. Dexamethasone was administered during the following 14-days and hemodynamic parameters were recorded at the end. Capillary density (CD) and capillary-to-fiber ratio (C:F ratio) were obtained in soleus muscle (SOL). Also, vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor-2 (VEGFR-2), endothelial nitric oxide synthase (eNOS), B-cell lymphoma 2 (Bcl-2), Bcl-2-like protein 4 (Bax), p-BAX and caspase-3 cleaved protein levels were analyzed. DEX treatment significantly increased blood pressure (+14%), which was associated with reduced C:F ratio (-41.0%) and CD (-43.1%). Reduction of vessel density was associated with decreased VEGF (-15.6%), VEGFR-2 (-14.6%), Bcl-2 (-18.4%), Bcl-2/Bax ratio (-29.0%) and p-Bax/Bax (-25.4%), and also with increased caspase-3 cleaved protein level (25%). Training, on the other hand, prevented microvessels loss by mitigating all proteins changes induced by DEX. In addition, angiogenic and apoptotic proteins were significantly correlated with CD, which, in turn, was associated with blood pressure. Therefore, we may point out that exercise training is a good strategy to attenuate DEX-induced microvascular rarefaction in soleus muscle and this response involves a better balance between apoptotic and angiogenic proteins, which may contribute for the attenuation of hypertension.


Subject(s)
Angiogenic Proteins/metabolism , Anti-Inflammatory Agents/adverse effects , Apoptosis Regulatory Proteins/metabolism , Dexamethasone/adverse effects , Microvascular Rarefaction/chemically induced , Physical Conditioning, Animal , Animals , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Male , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/physiopathology , Rats , Rats, Wistar
8.
Sci Rep ; 10(1): 440, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949240

ABSTRACT

Despite advances in renovascular disease (RVD) research, gaps remain between experimental and clinical outcomes, translation of results, and the understanding of pathophysiological mechanisms. A predictive tool to indicate support (or lack of) for biological findings may aid clinical translation of therapies. We created a Boolean model of RVD and hypothesized that it would predict outcomes observed in our previous studies using a translational swine model of RVD. Our studies have focused on developing treatments to halt renal microvascular (MV) rarefaction in RVD, a major feature of renal injury. A network topology of 20 factors involved in renal MV rarefaction that allowed simulation of 5 previously tested treatments was created. Each factor was assigned a function based upon its interactions with other variables and assumed to be "on" or "off". Simulations of interventions were performed until outcomes reached a steady state and analyzed to determine pathological processes that were activated, inactivated, or unchanged vs. RVD with no intervention. Boolean simulations mimicked the results of our previous studies, confirming the importance of MV integrity on treatment outcomes in RVD. Furthermore, our study supports the potential application of a mathematical tool to predict therapeutic feasibility, which may guide the design of future studies for RVD.


Subject(s)
Kidney Diseases/therapy , Microvascular Rarefaction/therapy , Models, Statistical , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Kidney Diseases/metabolism , Microvascular Rarefaction/metabolism , Swine , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
9.
Adv Wound Care (New Rochelle) ; 9(1): 1-8, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31871825

ABSTRACT

Objective: The healing of skin wounds is typified by a pattern of robust angiogenesis followed by vascular regression. Pigment epithelium-derived factor (PEDF), a recognized endogenous antiangiogenic protein, regulates vascular regression in resolving wounds through an unknown receptor. Among the multiple receptors for PEDF that have been identified, low-density lipoprotein receptor-related protein 6 (Lrp6) has been described as a regulator of angiogenesis in multiple systems. The purpose of the current study was to determine if the Lrp6 receptor plays a role in vessel regression in wounds. Approach: Excisional skin wounds were prepared on C57BL/6 mice. RT-PCR and immunoblots were performed to measure Lrp6 expression over a time course of wound healing. Immunohistochemistry was performed to localize Lrp6 in both recombinant PEDF (rPEDF)-treated and control wounds. To examine whether Lrp6 is critical to the regulation of capillary regression in vivo, wounds were treated with Lrp6 siRNA to minimize its presence in wounds. Immunohistochemistry for CD31 was performed to quantify blood vessel density. Results: PCR and immunoblots revealed significant increases in Lrp6 expression during the vascular regression phase of wound healing. Lrp6 was found to colocalize with CD31+ endothelial cells in wounds. The addition of rPEDF to wounds caused an increase in Lrp6-CD31+ endothelial cell colocalization. Inhibition of Lrp6 by siRNA impeded the vascular regression phase of healing. Innovation: This study is the first to demonstrate an association between Lrp6 and vessel regression in wound healing. Conclusion: Lrp6 is expressed in wounds in a temporal and spatial manner that suggests it may be a receptor for PEDF during vascular regression. PEDF increases Lrp6 expression in the wound vasculature, and inhibition of Lrp6 blocked vascular regression in wounds. The results suggest that Lrp6 is important to vascular regression in wounds, possibly through direct interaction with PEDF.


Subject(s)
Eye Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Microvascular Rarefaction/metabolism , Neovascularization, Pathologic/metabolism , Nerve Growth Factors/metabolism , Serpins/metabolism , Wound Healing/genetics , Animals , Capillaries/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Eye Proteins/administration & dosage , Female , Mice , Mice, Inbred C57BL , Nerve Growth Factors/administration & dosage , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Serpins/administration & dosage , Skin/blood supply , Skin/metabolism , Skin/pathology
10.
Mol Med Rep ; 19(4): 3168-3178, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816496

ABSTRACT

Loss of peritubular capillaries is a notable feature of progressive renal interstitial fibrosis. Astaxanthin (ASX) is a natural carotenoid with various biological activities. The present study aimed to evaluate the effect of ASX on unilateral ureteral obstruction (UUO)­induced renal fibrosis in mice. For that purpose, mice were randomly divided into five treatment groups: Sham, ASX 100 mg/kg, UUO, UUO + ASX 50 mg/kg and UUO + ASX 100 mg/kg. ASX was administered to the mice for 7 or 14 days following UUO. The results demonstrated that UUO­induced histopathological changes in the kidney tissue were prevented by ASX. Renal function was improved by ASX treatment, as evidenced by decreased blood urea nitrogen and serum creatinine levels. Furthermore, the extent of renal fibrosis and collagen deposition induced by UUO was suppressed by ASX. The levels of collagen I, fibronectin and α­smooth muscle actin were increased by UUO in mice or by transforming growth factor (TGF)­ß1 treatment in NRK­52E cells, and were reduced by ASX administration. In addition, ASX inhibited the UUO­induced decrease in peritubular capillary density by upregulating vascular endothelial growth factor and downregulating thrombospondin 1 levels. Inactivation of the TGF­ß1/Smad signaling pathway was involved in the anti­fibrotic mechanism of ASX in UUO mice and TGF­ß1­treated NRK­52E cells. In conclusion, ASX attenuated renal interstitial fibrosis and peritubular capillary rarefaction via inactivation of the TGF­ß1/Smad signaling pathway.


Subject(s)
Fibrinolytic Agents/pharmacology , Kidney Diseases/etiology , Kidney Diseases/pathology , Microvascular Rarefaction/etiology , Microvascular Rarefaction/pathology , Ureteral Obstruction/complications , Animals , Biomarkers , Biopsy , Cell Line , Disease Models, Animal , Fibrosis , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Male , Mice , Microvascular Rarefaction/drug therapy , Microvascular Rarefaction/metabolism , Rats , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xanthophylls/pharmacology
11.
EBioMedicine ; 42: 64-75, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30904607

ABSTRACT

BACKGROUND: Hypertensive patients exhibit decline in capillary density and endothelial progenitor cells (EPCs). However, whether capillary rarefaction in hypertension is associated with defect angiogenesis of EPCs remains unknown. We hypothesized that impaired mitochondrial function of late EPCs in hypertension is associated with the structural lack of capillary microcirculation via deficient CXCR4/JAK2/SIRT5 signaling. METHODS: We performed capillary microcirculation detection in hypertensive patients and healthy subjects. Angiogenic capacity and mitochondrial function of circulating EPCs were evaluated. The underlying mechanisms were further investigated by genetic inhibition and overexpression. FINDINGS: Capillary density of nail fold and eye fundus were significantly reduced in hypertensive patients, which was paralleled to decreased in vitro late EPC function and in vivo angiogenic capacity. Meanwhile the decline of EPC function in hypertension was accompanied by impaired mitochondrial ultrastructure, diminished mitochondrial membrane potential, reduced oxygen consumption, increased ROS generation and NADH level. Rotenone induced inhibition of oxygen consumption rate, excessive ROS generation and loss of MMP, which markedly decreased the in vitro functions of EPCs. Furthermore, SIRT5 expression of EPCs in hypertension was markedly reduced, which was correlated to mitochondrial dysfunction. CXCR4 gene transfer enhanced SIRT5 expression, improved mitochondrial functions and augmented angiogenic capacity of EPCs. The beneficial impacts of SIRT5 up-regulation on late EPC-mediated angiogenesis can be abrogated by blockade of CXCR4/JAK2/SIRT5 signaling pathway. INTERPRETATION: Mitochondrial dysfunction-mediated fall in angiogenic capacity due to deficient CXCR4/JAK2/SIRT5 signaling of late EPCs is probably responsible for the capillary rarefaction in hypertension. Our findings provide insight into the potential of EPC mitochondria as a novel target for the treatment of hypertension-related loss of microvascular density. FUNDS: National Nature Science Foundation of China, 973Program, the Nature Science Foundation of Guangdong.


Subject(s)
Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Janus Kinase 2/metabolism , Microvascular Rarefaction/metabolism , Mitochondria/metabolism , Neovascularization, Physiologic , Receptors, CXCR4/metabolism , Sirtuins/metabolism , Animals , Biomarkers , Disease Models, Animal , Gene Expression , Humans , Hypertension/etiology , Hypertension/metabolism , Hypertension/pathology , Janus Kinase 2/genetics , Male , Membrane Potential, Mitochondrial , Mice , Microvascular Rarefaction/diagnostic imaging , Microvascular Rarefaction/genetics , Mitochondria/genetics , Mitochondria/ultrastructure , Models, Biological , Neovascularization, Physiologic/genetics , Oxygen Consumption , Rats , Rats, Inbred SHR , Reactive Oxygen Species/metabolism , Receptors, CXCR4/genetics , Risk Factors , Signal Transduction , Sirtuins/genetics , Stem Cell Transplantation , Transduction, Genetic
12.
Front Immunol ; 9: 1130, 2018.
Article in English | MEDLINE | ID: mdl-29875776

ABSTRACT

Kidney transplantation entails a high likelihood of endothelial injury. The endothelium is a target of choice for injury by ischemia-reperfusion, alloantibodies, and autoantibodies. A certain degree of ischemia-reperfusion injury inevitably occurs in the immediate posttransplant setting and can manifest as delayed graft function. Acute rejection episodes, whether T-cell or antibody-mediated, can involve the graft micro- and macrovasculature, leading to endothelial injury and adverse long-term consequences on graft function and survival. In turn, caspase-3 activation in injured and dying endothelial cells favors the release of extracellular vesicles (apoptotic bodies and apoptotic exosome-like vesicles) that further enhance autoantibody production, complement deposition, and microvascular rarefaction. In this review, we present the evidence for endothelial injury, its causes and long-term consequences on graft outcomes in the field of kidney transplantation.


Subject(s)
Endothelium/metabolism , Kidney Transplantation/adverse effects , Animals , Autoimmunity , Endothelial Cells/metabolism , Endothelium/pathology , Endothelium/physiopathology , Graft Rejection/immunology , Graft Survival/immunology , Humans , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/pathology , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Vascular Remodeling
13.
J Cardiovasc Pharmacol ; 70(3): 194-201, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28678054

ABSTRACT

Dexamethasone (DEX) causes rarefaction. In contrast, training (T) prevents rarefaction and stimulates angiogenesis. This study investigated the mechanisms responsible for the preventive role of T in DEX-induced rarefaction. Rats underwent T or were kept sedentary (8 weeks) and were treated with DEX or saline during the following 14 days. Tibialis anterior muscle was used for measurements of capillary density (CD), capillary-to-fiber ratio (C:F ratio), superoxide dismutase CuZn (SOD-1), superoxide dismutase MnSOD (SOD-2), catalase (CAT) mRNA as well as SOD-1, SOD-2, CAT, vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor-2 (VEGF-R2), cyclooxygenase-2 (COX-2), B-cell lymphoma 2 (Bcl-2), Bd-2-like protein 4 (Bax), p-Bax, and caspase-3 cleaved protein levels. DEX decreased CD (-38.1%), C:F ratio (-30.0%), VEGF (-19.0%), VEGFR-2 (-20.1%), COX-2 (-22.8%), Bcl-2 (-20.5%), Bcl-2/Bax ratio (-13.7%), p-Bax/Bax (-20.0%) and increased SOD-2 (+41.6%) and caspase-3 cleaved (+24.1%). Conversely, T prevented reductions in CD (+54.2%), C:F ratio (+32.9%), VEGF (+25.3%), VEGFR-2 (+22.2%), COX-2 (+31.5%), Bcl-2 (+35.5%), Bcl-2/Bax ratio (+19.9%), p-Bax/Bax (+32.1%), and caspase-3 cleaved increase (-7.8%). T increased CAT mRNA (+21.5%) in the DEX-treated group. In conclusion, T prevented the DEX-induced rarefaction by increasing antioxidant enzymes resulting in a better balance between apoptotic and anti-apoptotic protein levels.


Subject(s)
Dexamethasone/toxicity , Microvascular Rarefaction/chemically induced , Microvascular Rarefaction/prevention & control , Physical Conditioning, Animal/physiology , Animals , Anti-Inflammatory Agents/toxicity , Antioxidants/metabolism , Male , Microvascular Rarefaction/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/methods , Rats , Rats, Wistar
14.
Curr Osteoporos Rep ; 15(1): 1-8, 2017 02.
Article in English | MEDLINE | ID: mdl-28110469

ABSTRACT

PURPOSE OF REVIEW: The objective of this literature review is to determine whether there are indications that microvascular complications occur in diabetic bone. Evidence definitively linking diabetic skeletal fragility with microvascular complications in bone remains elusive. RECENT FINDINGS: Circumstantial evidence, some recent and some lost to time, suggests that atherosclerotic vascular diseases such as peripheral arterial disease cause poor blood perfusion of bone and subsequent hypoxia and contribute to low bone density and high cortical porosity, patterns similar to some recently observed in diabetic subjects. Evidence also exists to suggest that potentially anti-angiogenic conditions, such as impaired vascular endothelial growth factor (VEGF) signaling, predominate in diabetic bone. Microvascular complications may contribute, in part, to diabetic skeletal fragility but data supporting this interpretation are primarily circumstantial at this time. This review highlights gaps in our knowledge and hopefully spurs further discussions and research on this topic.


Subject(s)
Bone Marrow/blood supply , Bone and Bones/blood supply , Diabetes Mellitus/epidemiology , Diabetic Angiopathies/epidemiology , Fractures, Bone/epidemiology , Microvascular Rarefaction/epidemiology , Bone and Bones/metabolism , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Fractures, Bone/metabolism , Fractures, Bone/physiopathology , Humans , Microvascular Rarefaction/metabolism , Microvascular Rarefaction/physiopathology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...