Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
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.
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
3.
Angiogenesis ; 27(1): 23-35, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37326760

ABSTRACT

Patients with chronic kidney disease (CKD) have an increased risk for cardiovascular morbidity and mortality. Capillary rarefaction may be both one of the causes as well as a consequence of CKD and cardiovascular disease. We reviewed the published literature on human biopsy studies and conclude that renal capillary rarefaction occurs independently of the cause of renal function decline. Moreover, glomerular hypertrophy may be an early sign of generalized endothelial dysfunction, while peritubular capillary loss occurs in advanced renal disease. Recent studies with non-invasive measurements show that capillary rarefaction is detected systemically (e.g., in the skin) in individuals with albuminuria, as sign of early CKD and/or generalized endothelial dysfunction. Decreased capillary density is found in omental fat, muscle and heart biopsies of patients with advanced CKD as well as in skin, fat, muscle, brain and heart biopsies of individuals with cardiovascular risk factors. No biopsy studies have yet been performed on capillary rarefaction in individuals with early CKD. At present it is unknown whether individuals with CKD and cardiovascular disease merely share the same risk factors for capillary rarefaction, or whether there is a causal relationship between rarefaction in renal and systemic capillaries. Further studies on renal and systemic capillary rarefaction, including their temporal relationship and underlying mechanisms are needed. This review stresses the importance of preserving and maintaining capillary integrity and homeostasis in the prevention and management of renal and cardiovascular disease.


Subject(s)
Cardiovascular Diseases , Microvascular Rarefaction , Renal Insufficiency, Chronic , Vascular Diseases , Humans , Capillaries/pathology , Cardiovascular Diseases/pathology , Microvascular Rarefaction/pathology , Kidney/pathology , Renal Insufficiency, Chronic/pathology , Vascular Diseases/pathology
4.
J Vet Intern Med ; 37(2): 556-566, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36807589

ABSTRACT

BACKGROUND: Hypoxia is a key driver of fibrosis and is associated with capillary rarefaction in humans. OBJECTIVES: Characterize capillary rarefaction in cats with chronic kidney disease (CKD). ANIMALS: Archival kidney tissue from 58 cats with CKD, 20 unaffected cats. METHODS: Cross-sectional study of paraffin-embedded kidney tissue utilizing CD31 immunohistochemistry to highlight vascular structures. Consecutive high-power fields from the cortex (10) and corticomedullary junction (5) were digitally photographed. An observer counted and colored the capillary area. Image analysis was used to determine the capillary number, average capillary size, and average percent capillary area in the cortex and corticomedullary junction. Histologic scoring was performed by a pathologist masked to clinical data. RESULTS: Percent capillary area (cortex) was significantly lower in CKD (median 3.2, range, 0.8-5.6) compared to unaffected cats (4.4, 1.8-7.0; P = <.001) and was negatively correlated with serum creatinine concentrations (r = -.36, P = .0013), glomerulosclerosis (r = -0.39, P = <.001), inflammation (r = -.30, P = .009), and fibrosis (r = -.30, P = .007). Capillary size (cortex) was significantly lower in CKD cats (2591 pixels, 1184-7289) compared to unaffected cats (4523 pixels, 1801-7618; P = <.001) and was negatively correlated with serum creatinine concentrations (r = -.40, P = <.001), glomerulosclerosis (r = -.44, P < .001), inflammation (r = -.42, P = <.001), and fibrosis (r = -.38, P = <.001). CONCLUSIONS AND CLINICAL IMPORTANCE: Capillary rarefaction (decrease in capillary size and percent capillary area) is present in kidneys of cats with CKD and is positively correlated with renal dysfunction and histopathologic lesions.


Subject(s)
Cat Diseases , Microvascular Rarefaction , Renal Insufficiency, Chronic , Humans , Cats , Animals , Microvascular Rarefaction/complications , Microvascular Rarefaction/pathology , Microvascular Rarefaction/veterinary , Cross-Sectional Studies , Creatinine , Kidney/pathology , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/veterinary , Fibrosis , Inflammation/pathology , Inflammation/veterinary , Cat Diseases/pathology
5.
Am J Physiol Renal Physiol ; 324(4): F374-F386, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36794755

ABSTRACT

Inflammation that develops with the release of chemokines and cytokines during acute kidney injury (AKI) has been shown to participate in functional renal recovery. Although a major research focus has been on the role of macrophages, the family of C-X-C motif chemokines that promote neutrophil adherence and activation also increases with kidney ischemia-reperfusion (I/R) injury. This study tested the hypothesis that intravenous delivery of endothelial cells (ECs) that overexpress (C-X-C motif) chemokine receptors 1 and 2 (CXCR1 and CXCR2, respectively) improves outcomes in kidney I/R injury. Overexpression of CXCR1/2 enhanced homing of endothelial cells to I/R-injured kidneys and limited interstitial fibrosis, capillary rarefaction, and tissue injury biomarkers (serum creatinine concentration and urinary kidney injury molecule-1) following AKI and also reduced expression of P-selectin and the rodent (C-X-C motif) chemokine cytokine-induced neutrophil chemoattractant (CINC)-2ß as well as the number of myeloperoxidase-positive cells in the postischemic kidney. The serum chemokine/cytokine profile, including CINC-1, showed similar reductions. These findings were not observed in rats given endothelial cells transduced with an empty adenoviral vector (null-ECs) or a vehicle alone. These data indicate that extrarenal endothelial cells that overexpress CXCR1 and CXCR2, but not null-ECs or vehicle alone, reduce I/R kidney injury and preserve kidney function in a rat model of AKI.NEW & NOTEWORTHY Inflammation facilitates kidney ischemia-reperfusion (I/R) injury. Endothelial cells (ECs) that were modified to overexpress (C-X-C motif) chemokine receptor (CXCR)1/2 (CXCR1/2-ECs) were injected immediately following kidney I/R injury. The interaction of CXCR1/2-ECs, but not ECs transduced with an empty adenoviral vector, with injured kidney tissue preserved kidney function and reduced production of inflammatory markers, capillary rarefaction, and interstitial fibrosis. The study highlights a functional role for the C-X-C chemokine pathway in kidney damage following I/R injury.


Subject(s)
Acute Kidney Injury , Microvascular Rarefaction , Reperfusion Injury , Rats , Animals , Endothelial Cells/metabolism , Microvascular Rarefaction/pathology , Acute Kidney Injury/pathology , Chemokines/metabolism , Inflammation/metabolism , Cytokines/metabolism , Kidney/metabolism , Receptors, Chemokine/metabolism , Fibrosis , Reperfusion Injury/pathology
6.
Am J Physiol Renal Physiol ; 324(1): F106-F123, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36395384

ABSTRACT

Vascular endothelial growth factor (VEGF) and its cognate receptor (VEGFR2) system are crucial for cell functions associated with angiogenesis and vasculogenesis. Klotho contributes to vascular health maintenance in the kidney and other organs in mammals, but it is unknown whether renoprotection by Klotho is dependent on VEGF/VEGFR2 signaling. We used heterozygous VEGFR2-haploinsufficient (VEGFR2+/-) mice resulting from heterozygous knockin of green fluorescent protein in the locus of fetal liver kinase 1 encoding VEGFR2 to test the interplay of Klotho, phosphate, and VEGFR2 in kidney function, the vasculature, and fibrosis. VEGFR2+/- mice displayed downregulated VEGF/VEGFR2 signaling in the kidney, lower density of peritubular capillaries, and accelerated kidney fibrosis, all of which were also found in the homozygous Klotho hypomorphic mice. High dietary phosphate induced higher plasma phosphate, greater peritubular capillary rarefaction, and more kidney fibrosis in VEGFR2+/- mice compared with wild-type mice. Genetic overexpression of Klotho significantly attenuated the elevated plasma phosphate, kidney dysfunction, peritubular capillary rarefaction, and kidney fibrosis induced by a high-phosphate diet in wild-type mice but only modestly ameliorated these changes in the VEGFR2+/- background. In cultured endothelial cells, VEGFR2 inhibition reduced free VEGFR2 but enhanced its costaining of an endothelial marker (CD31) and exacerbated phosphotoxicity. Klotho protein maintained VEGFR2 expression and attenuated high phosphate-induced cell injury, which was reduced by VEGFR2 inhibition. In conclusion, normal VEGFR2 function is required for vascular integrity and for Klotho to exert vascular protective and antifibrotic actions in the kidney partially through the regulation of VEGFR2 function.NEW & NOTEWORTHY This research paper studied the interplay of vascular endothelial growth factor receptor type 2 (VEGFR2), high dietary phosphate, and Klotho, an antiaging protein, in peritubular structure and kidney fibrosis. Klotho protein was shown to maintain VEGFR2 expression in the kidney and reduce high phosphate-induced cell injury. However, Klotho cytoprotection was attenuated by VEGFR2 inhibition. Thus, normal VEGFR2 function is required for vascular integrity and Klotho to exert vascular protective and antifibrotic actions in the kidney.


Subject(s)
Cytoprotection , Kidney Diseases , Kidney , Klotho Proteins , Microvascular Rarefaction , Vascular Endothelial Growth Factor Receptor-2 , Animals , Mice , Endothelial Cells/metabolism , Fibrosis , Kidney/blood supply , Kidney/pathology , Kidney Diseases/pathology , Microvascular Rarefaction/pathology , Phosphates/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/deficiency , Klotho Proteins/genetics , Klotho Proteins/metabolism
7.
J Cachexia Sarcopenia Muscle ; 13(6): 2712-2723, 2022 12.
Article in English | MEDLINE | ID: mdl-36102002

ABSTRACT

BACKGROUND: Muscle disuse from bed rest or spaceflight results in losses in muscle mass, strength and oxidative capacity. Capillary rarefaction may contribute to muscle atrophy and the reduction in oxidative capacity during bed rest. Artificial gravity may attenuate the negative effects of long-term space missions or bed rest. The aim of the present study was to assess (1) the effects of bed rest on muscle fibre size, fibre type composition, capillarization and oxidative capacity in the vastus lateralis and soleus muscles after 6 and 55 days of bed rest and (2) the effectiveness of artificial gravity in mitigating bed-rest-induced detriments to these parameters. METHODS: Nineteen participants were assigned to a control group (control, n = 6) or an intervention group undergoing 30 min of centrifugation (n = 13). All underwent 55 days of head-down tilt bed rest. Vastus lateralis and soleus biopsies were taken at baseline and after 6 and 55 days of bed rest. Fibre type composition, fibre cross-sectional area, capillarization indices and oxidative capacity were determined. RESULTS: After just 6 days of bed rest, fibre atrophy (-23.2 ± 12.4%, P < 0.001) and reductions in capillary-to-fibre ratio (C:F; 1.97 ± 0.57 vs. 1.56 ± 0.41, P < 0.001) were proportional in both muscles as reflected by a maintained capillary density. Fibre atrophy proceeded at a much slower rate between 6 and 55 days of bed rest (-11.6 ± 12.1% of 6 days, P = 0.032) and was accompanied by a 19.1% reduction in succinate dehydrogenase stain optical density (P < 0.001), without any further significant decrements in C:F (1.56 ± 0.41 vs. 1.49 ± 0.37, P = 0.459). Consequently, after 55 days of bed rest, the capillary supply-oxidative capacity ratio of a fibre had increased by 41.9% (P < 0.001), indicating a capillarization in relative excess of oxidative capacity. Even though the heterogeneity of capillary spacing (LogR SD) was increased after 55 days by 12.7% (P = 0.004), tissue oxygenation at maximal oxygen consumption of the fibres was improved after 55 days bed rest. Daily centrifugation failed to blunt the bed-rest-induced reductions in fibre size and oxidative capacity and capillary rarefaction. CONCLUSIONS: The relationship between fibre size and oxidative capacity with the capillary supply of a fibre is uncoupled during prolonged bed rest as reflected by a rapid loss of muscle mass and capillaries, followed at later stages by a more than proportional loss of mitochondria without further capillary loss. The resulting excessive capillary supply of the muscle after prolonged bed rest is advantageous for the delivery of substrates needed for subsequent muscle recovery.


Subject(s)
Microvascular Rarefaction , Humans , Microvascular Rarefaction/pathology , Bed Rest/adverse effects , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Muscle, Skeletal/pathology , Muscle Fibers, Skeletal/pathology
8.
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
9.
Heart Surg Forum ; 25(1): E042-E047, 2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35238310

ABSTRACT

BACKGROUND: Hypertrophic obstructive cardiomyopathy (HOCM) is a genetic cardiomyopathy characterized by microvascular ischemia and myocardial fibrosis. Microvessels play an important role in myocardial fibrosis in HOCM. However, the changes of myocardial microvessels and myocardial fibrosis in pediatric and adult patients with HOCM remain unclear. This study was to investigate the changes in myocardial microvessel density (MVD) and myocardial fibrosis in pediatric and adult patients with HOCM. METHODS: We analyzed the changes in MVD and myocardial fibrosis in myectomy left ventricular (LV) septal wall specimens in 12 adult patients and five pediatric patients with HOCM. Control myocardium from the LV septal wall was collected at autopsy of 5 adults and 4 pediatric individuals, who died of non-cardiac causes. RESULTS: There was no significant difference in MVD between pediatric HOCM patients and control subjects (706.4±187.5 vs. 940.2±491.1, P > 0.05), but the myocardial fibrosis area ratio was significantly increased in HOCM than in control subjects (10.6±3.5 vs. 4.9±1.2, P < 0.01). MVD was significantly reduced, and myocardial fibrosis area ratio was significantly higher in adult HOCM patients than in control subjects (i.e. 523.3± 209.4 vs. 845.7±260.7, P < 0.05; 12.8±5.1 vs. 4.4±1.3, P < 0.05). There was no significant difference in MVD and myocardial fibrosis between pediatric and adult HOCM patients (706.4±187.5 vs. 523.3±209.4, P > 0.05; 10.6±3.5 vs. 12.8±5.1, P > 0.05).  Conclusions: Pediatric and adult patients with HOCM have high myocardial fibrosis. The present findings suggest that myocardial microvascular density lesions contribute to myocardial fibrosis during childhood.


Subject(s)
Cardiomyopathy, Hypertrophic , Microvascular Rarefaction , Adult , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/diagnosis , Cardiomyopathy, Hypertrophic/pathology , Child , Fibrosis , Humans , Microvascular Rarefaction/pathology , Myocardium/pathology
10.
Am J Physiol Renal Physiol ; 321(3): F335-F351, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34338031

ABSTRACT

Ischemia-reperfusion injury (IRI) is a major risk factor for chronic renal failure. Caspase-3, an effector responsible for apoptosis execution, is activated within the peritubular capillary (PTC) in the early stage of IRI-induced acute kidney injury (AKI). Recently, we showed that caspase-3-dependent microvascular rarefaction plays a key role in fibrosis development after mild renal IRI. Here, we further characterized the role of caspase-3 in microvascular dysfunction and progressive renal failure in both mild and severe AKI, by performing unilateral renal artery clamping for 30/60 min with contralateral nephrectomy in wild-type (C57BL/6) or caspase-3-/- mice. In both forms of AKI, caspase-3-/- mice showed better long-term outcomes despite worse initial tubular injury. After 3 wk, they showed reduced PTC injury, decreased PTC collagen deposition and α-smooth muscle actin expression, and lower tubular injury scores compared with wild-type animals. Caspase-3-/- mice with severe IRI also showed better preservation of long-term renal function. Intravital imaging and microcomputed tomography revealed preserved PTC permeability and better terminal capillary density in caspase-3-/- mice. Collectively, these results demonstrate the pivotal importance of caspase-3 in regulating long-term renal function after IRI and establish the predominant role of PTC dysfunction as a major contributor to progressive renal dysfunction.NEW & NOTEWORTHY Our findings demonstrate the pivotal importance of caspase-3 in regulating renal microvascular dysfunction, fibrogenesis, and long-term renal impairment after acute kidney injury induced by ischemia-reperfusion injury. Furthermore, this study establishes the predominant role of peritubular capillary integrity as a major contributor to progressive renal dysfunction after ischemia-reperfusion injury.


Subject(s)
Acute Kidney Injury/metabolism , Caspase 3/metabolism , Renal Insufficiency, Chronic/metabolism , Reperfusion Injury/metabolism , Animals , Apoptosis/physiology , Capillaries/metabolism , Female , Kidney/metabolism , Mice, Inbred C57BL , Microvascular Rarefaction/pathology , Reperfusion Injury/pathology
11.
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
12.
Sci Rep ; 11(1): 1001, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441624

ABSTRACT

Microvascular disease and rarefaction are key pathological hallmarks of hypertension. The retina uniquely allows direct, non-invasive investigation of the microvasculature. Recently developed optical coherence tomography angiography now allows investigation of the fine retinal capillaries, which may provide a superior marker of overall vascular damage. This was a prospective cross-sectional study to collect retinal capillary density data on 300 normal eyes from 150 hypertensive adults, and to investigate possible associations with other organ damage markers. The average age of participants was 54 years and there was a greater proportion of males (85; 57%) than females. Multivariate, confounder adjusted linear regression showed that retinal capillary rarefaction in the parafovea was associated with increased pulse wave velocity (ß = - 0.4, P = 0.04), log-albumin/creatinine ratio (ß = - 0.71, P = 0.003), and with reduced estimated glomerular filtration rate (ß = 0.04, P = 0.02). Comparable significant associations were also found for whole-image vascular-density, for foveal vascular-density significant associations were found with pulse wave velocity and estimated glomerular filtration rate only. Our results indicate that retinal capillary rarefaction is associated with arterial stiffness and impaired kidney function. Retinal capillary rarefaction may represent a useful and simple test to assess the integrated burden of hypertension on the microvasculature irrespective of current blood pressure levels.


Subject(s)
Arteries/pathology , Capillaries/pathology , Fovea Centralis/pathology , Hypertension/pathology , Kidney/pathology , Microvascular Rarefaction/pathology , Retinal Vessels/pathology , Blood Pressure/physiology , Cross-Sectional Studies , Female , Fluorescein Angiography/methods , Glomerular Filtration Rate/physiology , Humans , Male , Microcirculation/physiology , Microvessels/pathology , Middle Aged , Prospective Studies , Pulse Wave Analysis/methods , Tomography, Optical Coherence/methods , Vascular Stiffness/physiology
13.
Am J Physiol Renal Physiol ; 317(5): F1383-F1397, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31509009

ABSTRACT

Acute kidney injury (AKI) is a strong independent predictor of mortality and often results in incomplete recovery of renal function, leading to progressive chronic kidney disease (CKD). Many clinical trials have been conducted on the basis of promising preclinical data, but no therapeutic interventions have been shown to improve long-term outcomes after AKI. This is partly due to the failure of preclinical studies to accurately model clinically relevant injury and long-term outcomes on CKD progression. Here, we evaluated the long-term effects of AKI on CKD progression in three animal models reflecting diverse etiologies of AKI: repeat-dose cisplatin, rhabdomyolysis, and ischemia-reperfusion injury. Using transdermal measurement of glomerular filtration rate as a clinically relevant measure of kidney function and quantification of peritubular capillary density to measure capillary rarefaction, we showed that repeat-dose cisplatin caused capillary rarefaction and decreased renal function in mice without a significant increase in interstitial fibrosis, whereas rhabdomyolysis-induced AKI led to severe interstitial fibrosis, but renal function and peritubular capillary density were preserved. Furthermore, long-term experiments in mice with unilateral ischemia-reperfusion injury showed that restoration of renal function 12 wk after a contralateral nephrectomy was associated with increasing fibrosis, but a reversal of capillary rarefaction was seen at 4 wk. These data demonstrate that clear dissociation between kidney function and fibrosis in these models of AKI to CKD progression and suggest that peritubular capillary rarefaction is more strongly associated with CKD progression than renal fibrosis.


Subject(s)
Acute Kidney Injury/etiology , Cisplatin/toxicity , Microvascular Rarefaction/pathology , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/complications , Rhabdomyolysis/complications , Animals , Antineoplastic Agents/toxicity , Fibrosis/etiology , Kidney/drug effects , Kidney/pathology , Male , Mice , Microvascular Rarefaction/etiology
14.
Sci Rep ; 9(1): 5909, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976042

ABSTRACT

The implantation of continuous - flow ventricular assist devices (VAD) is suggested to evoke angiodysplasia contributing to adverse events such as gastrointestinal bleeding. We evaluated in vivo capillary density and glycocalyx dimensions to investigate possible systemic microvascular changes in patients with chronic heart failure and VAD support vs. standard medical treatment. Forty-two patients with VAD support were compared to forty-one patients with ischemic and non-ischemic chronic heart failure (CHF) on standard pharmacotherapy and to a group of forty-two healthy subjects in a prospective cross-sectional study. Sublingual microcirculation was visualized using Sidestream Darkfield videomicroscopy and functional and perfused total capillary densities were quantified. Patients with VAD implantation were followed for one year and bleeding events were recorded. Median time after VAD implantation was 18 months. Patients were treated with centrifugal-flow devices (n = 31) or axial-flow devices (n = 11). Median functional capillary density was significantly lower in patients with VAD therapy as compared to CHF patients (196 vs. 255/mm2, p = 0.042, adjusted p-value). Functional and total capillary densities were 44% and 53% lower (both p < 0.001) in patients with VAD therapy when compared to healthy subjects. Cox regression analysis revealed loss of capillary density as a significant predictor of bleeding events during one -year follow-up of VAD patients (HR: 0.987, CI (95%): 0.977-0.998, p = 0.021 for functional and 0.992, CI (95%): 0.985-0.999, p = 0.03 for total capillary density). In conclusion, patients with VAD support exhibit capillary density rarefaction, which was associated with bleeding events. If confirmed independently, capillary impairment may be evaluated as novel marker of bleeding risk.


Subject(s)
Capillaries/pathology , Cardiomyopathies/therapy , Heart Failure/therapy , Heart-Assist Devices/adverse effects , Microvascular Rarefaction/etiology , Myocardial Ischemia/therapy , Aged , Cardiomyopathies/pathology , Case-Control Studies , Cross-Sectional Studies , Female , Follow-Up Studies , Heart Failure/pathology , Humans , Male , Microcirculation , Microvascular Rarefaction/pathology , Middle Aged , Myocardial Ischemia/pathology , Prognosis , Retrospective Studies
15.
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
16.
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
17.
Cardiology ; 141(4): 202-211, 2018.
Article in English | MEDLINE | ID: mdl-30820009

ABSTRACT

BACKGROUND: Hypertrophic obstructive cardiomyopathy (HOCM) is a myocardial disease characterized by fibrosis and microvascular ischemia. Microvessels play a critical role in myocardial fibrosis in HOCM. However, it remains unclear whether or not myocardial fibrosis is associated with microvascular density (MVD) changes. OBJECTIVE: The aim of the present study was to investigate whether a reduction in MVD is related to myocardial fibrosis in HOCM cardiac samples. METHODS: We analyzed MVD and fibrosis in myectomy left ventricular (LV) septal wall specimens from 53 HOCM patients. Control myocardium from the LV septal wall was collected at autopsy of 9 individuals who died of noncardiac causes. RESULTS: The fibrosis ratio (% area) in HOCM was higher and the MVD was lower than that in control subjects (i.e., 12.7 ± 10.0 vs. 4.0 ± 1.4%, p = 0.012, and 480.9 ± 206.7 vs. 1,425 ± 221/mm2, p < 0.001). Patients with mild fibrosis had a higher MVD than patients with moderate fibrosis (i.e., 568.2 ± 214.8 vs. 403.2 ± 167.8/mm2, p = 0.006) and patients with severe fibrosis (i.e., 568.2 ± 214.8 vs. 378.6 ± 154.0/mm2, p = 0.024). Furthermore, a significant negative correlation was found between myocardial fibrosis and MVD in HOCM patients (r = -0.40, p = 0.003), which was also found in mild fibrosis (r = -0.40, p = 0.043), moderate fibrosis (r = -0.50, p = 0.024), and severe fibrosis (r = -0.24, p = 0.61), although no significant differences were observed in severe fibrosis. Additionally, we demonstrated that late gadolinium enhancement was negatively correlated with MVD (r = -0.37, p = 0.03) and positively correlated with fibrosis (r = 0.44, p = 0.01). CONCLUSION: HOCM patients had a higher myocardial fibrosis ratio and a lower MVD. The severity of myocardial fibrosis was negatively correlated with MVD in HOCM. These findings showed that a reduced MVD may contribute to myocardial fibrosis in HOCM.


Subject(s)
Cardiomyopathy, Hypertrophic/diagnostic imaging , Heart Septum/diagnostic imaging , Magnetic Resonance Imaging, Cine , Microvascular Rarefaction/diagnostic imaging , Myocardium/pathology , Adult , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/pathology , Contrast Media , Echocardiography , Female , Fibrosis , Gadolinium DTPA , Heart Septum/pathology , Humans , Male , Microvascular Rarefaction/etiology , Microvascular Rarefaction/pathology , Middle Aged , Myocardial Contraction
18.
Pediatr Nephrol ; 32(7): 1275-1278, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28435991

ABSTRACT

BACKGROUND: Low birthweight infants have a reduced number of nephrons and are at high risk of chronic kidney disease. Preterm birth and/or intrauterine growth restriction (IUGR) may also affect peritubular capillary development, as has been shown in other organs. CASE-DIAGNOSIS/TREATMENT: We report two patients with a history of preterm birth and extremely low birthweight who showed polycythemia and renal capillary rarefaction. Patient 1 and 2, born at 25 weeks of gestation with a birthweight of 728 and 466 g, showed mild proteinuria at age 8 and 6 years, respectively. In addition to increasing proteinuria, hemoglobin levels became elevated towards adolescence and their serum erythropoietin (EPO) was high despite polycythemia. Light microscopic examination of renal biopsy specimens showed glomerular hypertrophy, focal segmental glomerulosclerosis, and only mild tubulointerstitial fibrosis. A decrease in the immunohistochemical staining of CD31 and CD34 endothelial cells in renal biopsy specimens was consistent with peritubular capillary rarefaction. CONCLUSIONS: Since kidney function was almost normal and fibrosis was not severe, we consider that the capillary rarefaction and polycythemia associated with elevated EPO levels were largely attributable to preterm birth and/or IUGR.


Subject(s)
Glomerulosclerosis, Focal Segmental/pathology , Infant, Premature, Diseases/pathology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Microvascular Rarefaction/pathology , Nephrons/pathology , Polycythemia/pathology , Premature Birth/pathology , Adolescent , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antigens, CD34 , Apgar Score , Biopsy , Child , Endothelial Cells/metabolism , Erythropoietin/blood , Female , Fibrosis , Glomerulosclerosis, Focal Segmental/diagnosis , Glomerulosclerosis, Focal Segmental/therapy , Glomerulosclerosis, Focal Segmental/urine , Hemoglobins/analysis , Humans , Infant, Extremely Premature , Infant, Newborn , Infant, Premature, Diseases/diagnosis , Infant, Premature, Diseases/therapy , Infant, Premature, Diseases/urine , Infant, Very Low Birth Weight , Male , Microvascular Rarefaction/blood , Microvascular Rarefaction/diagnosis , Microvascular Rarefaction/therapy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Polycythemia/blood , Polycythemia/diagnosis , Polycythemia/urine , Pregnancy , Proteinuria/urine , Valsartan/therapeutic use
19.
Sci Rep ; 7: 40156, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071761

ABSTRACT

Severe hypertension can lead to malignant hypertension (MH) with renal thrombotic microangiopathy and hemolysis. The role of plasma heme release in this setting is unknown. We aimed at evaluating the effect of a mild plasma heme increase by hemin administration in angiotensin II (AngII)-mediated hypertensive rats. Prevalence of MH and blood pressure values were similar in AngII and AngII + hemin groups. MH rats displayed a decreased renal blood flow (RBF), increased renal vascular resistances (RVR), and increased aorta and interlobar arteries remodeling with a severe renal microcirculation assessed by peritubular capillaries (PTC) rarefaction. Hemin-treated rats with or without AngII displayed also a decreased RBF and increased RVR explained only by PCT rarefaction. In AngII rats, RBF was similar to controls (with increased RVR). PTC density appeared strongly correlated to tubular damage score (rho = -0.65, p < 0.0001) and also renal Heme Oygenase-1 (HO-1) mRNA (rho = -0.67, p < 0.0001). HO-1 was expressed in PTC and renal tubules in MH rats, but only in PTC in other groups. In conclusion, though increased plasma heme does not play a role in triggering or aggravating MH, heme release appears as a relevant toxic mediator leading to renal impairment, primarily through PTC endothelial dysfunction rather than direct tubular toxicity.


Subject(s)
Heme/toxicity , Hypertension/complications , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Microvascular Rarefaction/pathology , Plasma/chemistry , Animals , Hemin/administration & dosage , Rats, Sprague-Dawley , Renal Circulation
20.
Age (Dordr) ; 38(4): 273-289, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27613724

ABSTRACT

Strong epidemiological and experimental evidence indicate that both age and hypertension lead to significant functional and structural impairment of the cerebral microcirculation, predisposing to the development of vascular cognitive impairment (VCI) and Alzheimer's disease. Preclinical studies establish a causal link between cognitive decline and microvascular rarefaction in the hippocampus, an area of brain important for learning and memory. Age-related decline in circulating IGF-1 levels results in functional impairment of the cerebral microvessels; however, the mechanistic role of IGF-1 deficiency in impaired hippocampal microvascularization remains elusive. The present study was designed to characterize the additive/synergistic effects of IGF-1 deficiency and hypertension on microvascular density and expression of genes involved in angiogenesis and microvascular regression in the hippocampus. To achieve that goal, we induced hypertension in control and IGF-1 deficient mice (Igf1 f/f  + TBG-Cre-AAV8) by chronic infusion of angiotensin II. We found that circulating IGF-1 deficiency is associated with decreased microvascular density and exacerbates hypertension-induced microvascular rarefaction both in the hippocampus and the neocortex. The anti-angiogenic hippocampal gene expression signature observed in hypertensive IGF-1 deficient mice in the present study provides important clues for subsequent studies to elucidate mechanisms by which hypertension may contribute to the pathogenesis and clinical manifestation of VCI. In conclusion, adult-onset, isolated endocrine IGF-1 deficiency exerts deleterious effects on the cerebral microcirculation, leading to a significant decline in cortical and hippocampal capillarity and exacerbating hypertension-induced cerebromicrovascular rarefaction. The morphological impairment of the cerebral microvasculature induced by IGF-1 deficiency and hypertension reported here, in combination with neurovascular uncoupling, increased blood-brain barrier disruption and neuroinflammation reported in previous studies likely contribute to the pathogenesis of vascular cognitive impairment in elderly hypertensive humans.


Subject(s)
Aging/metabolism , Hippocampus/blood supply , Hypertension/complications , Insulin-Like Growth Factor I/deficiency , Microvascular Rarefaction/pathology , Neocortex/blood supply , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Angiotensin II/adverse effects , Angiotensin II/metabolism , Animals , Biomarkers/blood , Blood-Brain Barrier/metabolism , Cognitive Dysfunction/physiopathology , Gene Expression , Humans , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Inbred C57BL , Microvascular Rarefaction/etiology , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...