Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
1.
Am J Physiol Renal Physiol ; 326(2): F257-F264, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38031731

ABSTRACT

Renal artery stenosis (RAS) is a major cause of ischemic kidney disease, which is largely mediated by inflammation. Mapping the immune cell composition in ischemic kidneys might provide useful insight into the disease pathogenesis and uncover therapeutic targets. We used mass cytometry (CyTOF) to explore the single-cell composition in a unique data set of human kidneys nephrectomized due to chronic occlusive vascular disease (RAS, n = 3), relatively healthy donor kidneys (n = 6), and unaffected sections of kidneys with renal cell carcinoma (RCC, n = 3). Renal fibrosis and certain macrophage populations were also evaluated in renal sections. Cytobank analysis showed in RAS kidneys decreased cell populations expressing epithelial markers (CD45-/CD13+) and increased CD45+ inflammatory cells, whereas scattered tubular-progenitor-like cells (CD45-/CD133+/CD24+) increased compared with kidney donors. Macrophages switched to proinflammatory phenotypes in RAS, and the numbers of IL-10-producing dendritic cells (DC) were also lower. Compared with kidney donors, RAS kidneys had decreased overall DC populations but increased plasmacytoid DC. Furthermore, senescent active T cells (CD45+/CD28+/CD57+), aged neutrophils (CD45+/CD15+/CD24+/CD11c+), and regulatory B cells (CD45+/CD14-/CD24+/CD44+) were increased in RAS. RCC kidneys showed a distribution of cell phenotypes comparable with RAS but less pronounced, accompanied by an increase in CD34+, CD370+, CD103+, and CD11c+/CD103+ cells. Histologically, RAS kidneys showed significantly increased fibrosis and decreased CD163+/CD141+ cells. The single-cell platform CyTOF enables the detection of significant changes in renal cells, especially in subsets of immune cells in ischemic human kidneys. Endogenous pro-repair cell types in RAS warrant future study for potential immune therapy.NEW & NOTEWORTHY The single-cell platform mass cytometry (CyTOF) enables detection of significant changes in one million of renal cells, especially in subsets of immune cells in ischemic human kidneys distal to renal artery stenosis (RAS). We found that pro-repair cell types such as scattered tubular-progenitor-like cells, aged neutrophils, and regulatory B cells show a compensatory increase in RAS. Immune cell phenotype changes may reflect ongoing inflammation and impaired immune defense capability in the kidneys.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Renal Artery Obstruction , Humans , Aged , Carcinoma, Renal Cell/pathology , Renal Artery Obstruction/pathology , Renal Artery , Kidney/pathology , Ischemia/pathology , Phenotype , Inflammation/pathology , Kidney Neoplasms/pathology
2.
Stem Cells ; 41(1): 50-63, 2023 01 30.
Article in English | MEDLINE | ID: mdl-36250949

ABSTRACT

Atherosclerotic renal artery stenosis (ARAS) is associated with irreversible parenchymal renal disease and regenerative stem cell therapies may improve renal outcomes. Hypoxia preconditioning (HPC) may improve the regenerative functions of adipose tissue-derived mesenchymal stem cells (AMSC) by affecting DNA 5-hydroxymethylcytosine (5hmC) marks in angiogenic genes. Here, we investigated using a porcine ARAS model, whether growth of ARAS AMSCs in hypoxia (Hx) versus normoxia (Nx) would enhance renal tissue repair, and comprehensively analyze how HPC modifies DNA hydroxymethylation compared to untreated ARAS and healthy/normal pigs (n=5 each). ARAS pigs exhibited elevated serum cholesterol, serum creatinine and renal artery stenosis, with a concomitant decrease in renal blood flow (RBF) and increased blood pressure (BP) compared to healthy pigs. Renal artery injection of either autologous Nx or Hx AMSCs improved diastolic BP, reduced kidney tissue fibrosis, and inflammation (CD3+ T-cells) in ARAS pigs. In addition, renal medullary hypoxia significantly lowered with Nx but not Hx AMSC treatment. Mechanistically, levels of epigenetic 5hmC marks (which reflect gene activation) estimated using DNA immunoprecipitation technique were elevated in profibrotic and inflammatory genes in ARAS compared with normal AMSCs. HPC significantly reduced 5hmC levels in cholesterol biosynthesis and oxidative stress response pathways in ARAS AMSCs. Thus, autologous AMSCs improve key renovascular parameters and inflammation in ARAS pigs, with HPC mitigating pathological molecular effects on inflammatory and profibrotic genes which may play a role in augmenting regenerative capacity of AMSCs.


Subject(s)
Mesenchymal Stem Cells , Renal Artery Obstruction , Swine , Animals , Renal Artery Obstruction/therapy , Renal Artery Obstruction/pathology , Hypoxia/metabolism , Mesenchymal Stem Cells/metabolism , Cholesterol/metabolism , Inflammation/pathology , Adipose Tissue/metabolism
3.
J Hypertens ; 40(10): 1859-1875, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36052518

ABSTRACT

Even though it has been more than a decade since renal denervation (RDN) was first used to treat hypertension and an intense effort on researching this therapy has been made, it is still not clear how RDN fits into the antihypertensive arsenal. There is no question that RDN lowers blood pressure (BP), it does so to an extent at best corresponding to one antihypertensive drug. The procedure has an excellent safety record. However, it remains clinically impossible to predict whose BP responds to RDN and whose does not. Long-term efficacy data on BP reduction are still unconvincing despite the recent results in the SPYRAL HTN-ON MED trial; experimental studies indicate that reinnervation is occurring after RDN. Although BP is an acceptable surrogate endpoint, there is complete lack of outcome data with RDN. Clear indications for RDN are lacking although patients with resistant hypertension, those with documented increase in activity of the sympathetic system and perhaps those who desire to take fewest medication may be considered.


Subject(s)
Antihypertensive Agents , Hypertension , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Blood Pressure , Denervation/methods , Humans , Hypertension/drug therapy , Hypertension/surgery , Kidney , Sympathectomy/methods , Treatment Outcome
4.
Hypertension ; 79(8): e128-e143, 2022 08.
Article in English | MEDLINE | ID: mdl-35708012

ABSTRACT

Renovascular disease is a major causal factor for secondary hypertension and renal ischemic disease. However, several prospective, randomized trials for atherosclerotic disease failed to demonstrate that renal revascularization is more effective than medical therapy for most patients. These results have greatly reduced the generalized diagnostic workup and use of renal revascularization. Most guidelines and review articles emphasize the limited average improvement and fail to identify those clinical populations that do benefit from revascularization. On the basis of the clinical experience of hypertension centers, specialists have continued selective revascularization, albeit without a summary statement by a major, multidisciplinary, national organization that identifies specific populations that may benefit. In this scientific statement for health care professionals and the public-at-large, we review the strengths and weaknesses of randomized trials in revascularization and highlight (1) when referral for consideration of diagnostic workup and therapy may be warranted, (2) the evidence/rationale for these selective scenarios, (3) interventional and surgical techniques for effective revascularization, and (4) areas of research with unmet need.


Subject(s)
Hypertension, Renovascular , Hypertension , Renal Artery Obstruction , American Heart Association , Humans , Hypertension, Renovascular/diagnosis , Hypertension, Renovascular/etiology , Hypertension, Renovascular/surgery , Prospective Studies , Renal Artery Obstruction/diagnosis , Renal Artery Obstruction/surgery , Vascular Surgical Procedures
5.
Nephrol Dial Transplant ; 37(10): 1844-1856, 2022 09 22.
Article in English | MEDLINE | ID: mdl-35451482

ABSTRACT

BACKGROUND: Renal artery stenosis (RAS) is an important cause of chronic kidney disease and secondary hypertension. In animal models, renal ischemia leads to downregulation of growth factor expression and loss of intrarenal microcirculation. However, little is known about the sequelae of large-vessel occlusive disease on the microcirculation within human kidneys. METHOD: This study included five patients who underwent nephrectomy due to renovascular occlusion and seven nonstenotic discarded donor kidneys (four deceased donors). Micro-computed tomography was performed to assess microvascular spatial densities and tortuosity, an index of microvascular immaturity. Renal protein expression, gene expression and histology were studied in vitro using immunoblotting, polymerase chain reaction and staining. RESULTS: RAS demonstrated a loss of medium-sized vessels (0.2-0.3 mm) compared with donor kidneys (P = 0.037) and increased microvascular tortuosity. RAS kidneys had greater protein expression of angiopoietin-1, hypoxia-inducible factor-1α and thrombospondin-1 but lower protein expression of vascular endothelial growth factor (VEGF) than donor kidneys. Renal fibrosis, loss of peritubular capillaries (PTCs) and pericyte detachment were greater in RAS, yet they had more newly formed PTCs than donor kidneys. Therefore, our study quantified significant microvascular remodeling in the poststenotic human kidney. RAS induced renal microvascular loss, vascular remodeling and fibrosis. Despite downregulated VEGF, stenotic kidneys upregulated compensatory angiogenic pathways related to angiopoietin-1. CONCLUSIONS: These observations underscore the nature of human RAS as a microvascular disease distal to main vessel stenosis and support therapeutic strategies directly targeting the poststenotic kidney microcirculation in patients with RAS.


Subject(s)
Renal Artery Obstruction , Angiopoietin-1/metabolism , Angiopoietin-1/therapeutic use , Animals , Fibrosis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Renal Artery Obstruction/complications , Renal Circulation/physiology , Thrombospondins/metabolism , Vascular Endothelial Growth Factor A/metabolism , X-Ray Microtomography
6.
Am J Kidney Dis ; 79(2): 289-301, 2022 02.
Article in English | MEDLINE | ID: mdl-34384806

ABSTRACT

The diagnosis and management of atherosclerotic renovascular disease (ARVD) is complex and controversial. Despite evidence from the ASTRAL (2009) and CORAL (2013) randomized controlled trials showing that percutaneous renal artery revascularization did not improve major outcomes compared with best medical therapy alone over 3-5 years, several areas of uncertainty remain. Medical therapy, including statin and antihypertensive medications, has evolved in recent years, and the use of renin-angiotensin-aldosterone system blockers is now considered the primary means to treat hypertension in the setting of ARVD. However, the criteria to identify kidneys with renal artery stenosis that have potentially salvageable function are evolving. There are also data suggesting that certain high-risk populations with specific clinical manifestations may benefit from revascularization. Here, we provide an overview of the epidemiology, diagnosis, and treatment of ARVD based on consensus recommendations from a panel of physician experts who attended the recent KDIGO (Kidney Disease: Improving Global Outcomes) Controversies Conference on central and peripheral arterial diseases in chronic kidney disease. Most focus is provided for contentious issues, and we also outline aspects of investigation and management of ARVD that require further research.


Subject(s)
Atherosclerosis , Hypertension, Renovascular , Renal Artery Obstruction , Atherosclerosis/diagnosis , Atherosclerosis/epidemiology , Atherosclerosis/therapy , Humans , Hypertension, Renovascular/diagnosis , Hypertension, Renovascular/epidemiology , Hypertension, Renovascular/etiology , Kidney , Renal Artery , Renal Artery Obstruction/diagnosis , Renal Artery Obstruction/epidemiology , Renal Artery Obstruction/therapy , Renin-Angiotensin System
7.
J Am Soc Nephrol ; 32(8): 1987-2004, 2021 08.
Article in English | MEDLINE | ID: mdl-34135081

ABSTRACT

BACKGROUND: Peripheral vascular diseases may induce chronic ischemia and cellular injury distal to the arterial obstruction. Cellular senescence involves proliferation arrest in response to stress, which can damage neighboring cells. Renal artery stenosis (RAS) induces stenotic-kidney dysfunction and injury, but whether these arise from cellular senescenceand their temporal pattern remain unknown. METHODS: Chronic renal ischemia was induced in transgenic INK-ATTAC and wild type C57BL/6 mice by unilateral RAS, and kidney function (in vivo micro-MRI) and tissue damage were assessed. Mouse healthy and stenotic kidneys were analyzed using unbiased single-cell RNA-sequencing. To demonstrate translational relevance, cellular senescence was studied in human stenotic kidneys. RESULTS: Using intraperitoneal AP20187 injections starting 1, 2, or 4 weeks after RAS, selective clearance of cells highly expressing p16Ink4a attenuated cellular senescence and improved stenotic-kidney function; however, starting treatment immediately after RAS induction was unsuccessful. Broader clearance of senescent cells, using the oral senolytic combination dasatinib and quercetin, in C57BL/6 RAS mice was more effective in clearing cells positive for p21 (Cdkn1a) and alleviating renal dysfunction and damage. Unbiased, single-cell RNA sequencing in freshly dissociated cells from healthy and stenotic mouse kidneys identified stenotic-kidney epithelial cells undergoing both mesenchymal transition and senescence. As in mice, injured human stenotic kidneys exhibited cellular senescence, suggesting this process is conserved. CONCLUSIONS: Maladaptive tubular cell senescence, involving upregulated p16 (Cdkn2a), p19 (Cdkn2d), and p21 (Cdkn1a) expression, is associated with renal dysfunction and injury in chronic ischemia. These findings support development of senolytic strategies to delay chronic ischemic renal injury.


Subject(s)
Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Ischemia/physiopathology , Kidney/physiopathology , Renal Insufficiency, Chronic/physiopathology , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Caspase 8/metabolism , Cellular Senescence/drug effects , Cellular Senescence/genetics , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Dasatinib/pharmacology , Disease Models, Animal , Enzyme Activation/drug effects , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Gene Expression , Heparin-binding EGF-like Growth Factor/genetics , Humans , Ischemia/etiology , Kidney/blood supply , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteopontin/genetics , Protein Kinase Inhibitors/pharmacology , Renal Artery Obstruction/complications , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology , Sequence Analysis, RNA , Single-Cell Analysis , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Up-Regulation , p21-Activated Kinases/genetics
8.
Cells ; 10(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33807289

ABSTRACT

Ischemic nephropathy reflects progressive loss of kidney function due to large vessel atherosclerotic occlusive disease. Recent studies indicate that this process is characterized by microvascular rarefaction, increased tissue hypoxia and activation of inflammatory processes of tissue injury. This review summarizes the rationale and application of functional MR imaging to evaluate tissue oxygenation in human subjects that defines the limits of renal adaptation to reduction in blood flow, development of increasingly severe tissue hypoxia and recruitment of inflammatory injury pathways in ischemic nephropathy. Human mesenchymal stromal/stem cells (MSC) are capable of modifying angiogenic pathways and immune responses, but the potency of these effects vary between individuals and various clinical characteristics including age and chronic kidney disease and levels of hypoxia. We summarize recently completed first-in-human studies applying intrarenal infusion of autologous adipose-derived MSC in human subjects with ischemic nephropathy that demonstrate a rise in blood flow and reduction in tissue hypoxia consistent with partial repair of microvascular injury, even without restoring main renal arterial blood flow. Inflammatory biomarkers in the renal vein of post-stenotic kidneys fell after MSC infusion. These changes were associated with modest but significant dose-related increments in kidney function. These data provide support a role for autologous MSC in repair of microvascular injury associated with tissue hypoxia.


Subject(s)
Ischemia/therapy , Kidney Diseases/therapy , Kidney/blood supply , Mesenchymal Stem Cell Transplantation , Microvessels/injuries , Microvessels/pathology , Animals , Humans , Hypoxia/complications , Hypoxia/pathology , Hypoxia/therapy , Ischemia/complications , Ischemia/pathology , Kidney Diseases/complications , Kidney Diseases/pathology
9.
Stem Cell Res Ther ; 12(1): 240, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33853680

ABSTRACT

BACKGROUND: Atherosclerotic renal artery stenosis (ARAS) is a risk factor for ischemic and hypertensive kidney disease (HKD) for which autologous mesenchymal stem cell (MSC) appears to be a promising therapy. However, MSCs from ARAS patients exhibit impaired function, senescence, and DNA damage, possibly due to epigenetic mechanisms. Hypoxia preconditioning (HPC) exerts beneficial effects on cellular proliferation, differentiation, and gene and protein expression. We hypothesized that HPC could influence MSC function and senescence in ARAS by epigenetic mechanisms and modulating gene expression of chromatin-modifying enzymes. METHODS: Adipose-derived MSC harvested from healthy control (N = 8) and ARAS (N = 8) pigs were cultured under normoxia (20%O2) or hypoxia (1%O2) conditions. MSC function was assessed by migration, proliferation, and cytokine release in conditioned media. MSC senescence was evaluated by SA-ß-gal activity. Specific pro-angiogenic and senescence genes were assessed by reverse transcription polymerase chain reaction (RT-PCR). Dot blotting was used to measure global genome 5-hydroxymethylcytosine (5hmC) levels on DNA and Western blotting of modified histone 3 (H3) proteins to quantify tri-methylated lysine-4 (H3K4me3), lysine-9 (H3K9me3), and lysine-27 (H3K27me3) residues. RESULTS: Specific pro-angiogenic genes in ARAS assessed by RT-PCR were lower at baseline but increased under HPC, while pro-senescence genes were higher in ARAS at baseline as compared healthy MSCs. ARAS MSCs under basal conditions, displayed higher H3K4me3, H3K27me3, and 5hmC levels compared to healthy MSCs. During HPC, global 5hmC levels were decreased while no appreciable changes occurred in histone H3 tri-methylation. ARAS MSCs cultured under HPC had higher migratory and proliferative capacity as well as increased vascular endothelial growth factor and epidermal growth factor expression compared to normoxia, and SA-ß-gal activity decreased in both animal groups. CONCLUSIONS: These data demonstrate that swine ARAS MSCs have decreased angiogenesis and increased senescence compared to healthy MSCs and that HPC mitigates MSC dysfunction, senescence, and DNA hydroxymethylation in ARAS MSC. Thus, HPC for MSCs may be considered for their optimization to improve autologous cell therapy in patients with nephropathies.


Subject(s)
Mesenchymal Stem Cells , Renal Artery Obstruction , Animals , Cells, Cultured , Epigenesis, Genetic , Humans , Hypoxia , Swine , Vascular Endothelial Growth Factor A
10.
Diabetes ; 70(7): 1561-1574, 2021 07.
Article in English | MEDLINE | ID: mdl-33858824

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) facilitate repair in experimental diabetic kidney disease (DKD). However, the hyperglycemic and uremic milieu may diminish regenerative capacity of patient-derived therapy. We hypothesized that DKD reduces human MSC paracrine function. Adipose-derived MSC from 38 participants with DKD and 16 control subjects were assessed for cell surface markers, trilineage differentiation, RNA sequencing (RNA-seq), in vitro function (coculture or conditioned medium experiments with T cells and human kidney cells [HK-2]), secretome profile, and cellular senescence abundance. The direction of association between MSC function and patient characteristics were also tested. RNA-seq analysis identified 353 differentially expressed genes and downregulation of several immunomodulatory genes/pathways in DKD-MSC versus Control-MSC. DKD-MSC phenotype, differentiation, and tube formation capacity were preserved, but migration was reduced. DKD-MSC with and without interferon-γ priming inhibited T-cell proliferation greater than Control-MSC. DKD-MSC medium contained higher levels of anti-inflammatory cytokines (indoleamine 2,3-deoxygenase 1 and prostaglandin-E2) and prorepair factors (hepatocyte growth factor and stromal cell-derived factor 1) but lower IL-6 versus control-MSC medium. DKD-MSC medium protected high glucose plus transforming growth factor-ß-exposed HK-2 cells by reducing apoptotic, fibrotic, and inflammatory marker expression. Few DKD-MSC functions were affected by patient characteristics, including age, sex, BMI, hemoglobin A1c, kidney function, and urine albumin excretion. However, senescence-associated ß-galactosidase activity was lower in DKD-MSC from participants on metformin therapy. Therefore, while DKD altered the transcriptome and migratory function of culture-expanded MSCs, DKD-MSC functionality, trophic factor secretion, and immunomodulatory activities contributing to repair remained intact. These observations support testing of patient-derived MSC therapy and may inform preconditioning regimens in DKD clinical trials.


Subject(s)
Adipose Tissue/cytology , Diabetic Nephropathies/physiopathology , Immunomodulation , Mesenchymal Stem Cells/physiology , Transcriptome , Apoptosis , Cells, Cultured , Cellular Senescence , Diabetic Nephropathies/immunology , Humans , Lymphocyte Activation , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology
11.
Clin Transplant ; 35(6): e14293, 2021 06.
Article in English | MEDLINE | ID: mdl-33745214

ABSTRACT

BACKGROUND: The medium- to long-term outcomes of living kidney donors with hypertension compared to normotensive donors are not well understood, especially with the recent changes in hypertension guidelines. METHODS: We studied a cohort of 950 living kidney donors using different definitions of hypertension based on either ≥140/90 or ≥130/80 mmHg thresholds and based on either office or ambulatory blood pressure readings. Microstructural features on kidney biopsy at the time of donation were compared using different definitions of hypertension. RESULTS: After adjusting for years of follow-up, age, sex, and baseline eGFR, hypertension (by any definition) did not significantly predict an eGFR < 45 ml/min/1.73 m2 at a median follow-up of 10 years postdonation, though there was a borderline association with ambulatory blood pressure ≥ 130/80 mmHg predicting a 40% decline in eGFR (OR = 1.53, 1.00-2.36; p = .051). Proteinuria was predicted by office blood pressure ≥ 140/90 mmHg and by nondipper profile on nocturnal ambulatory blood pressure measurements. At the time of donation, larger glomeruli and arterial hyalinosis on biopsy were associated with hypertension defined by either ≥140/90 or ≥130/80 mmHg (by office or ambulatory measurements). Nocturnal nondipper status was associated with larger glomeruli size but not arteriolar hyalinosis when compared to dippers. CONCLUSIONS: In programs that accept donors with controlled hypertension, various definitions of hypertension are associated with histological findings in the donated kidney, but none predict a clinically significant decline in kidney function 10 years after donation. These data support allowing healthy individuals with controlled hypertension to donate a kidney. However, donors with office hypertension (≥140/90 mmHg) and nondippers (regardless of hypertension status) are at greater long-term risk for proteinuria, and particularly for these donors, longer follow-up is warranted.


Subject(s)
Hypertension , Kidney Transplantation , Biopsy , Blood Pressure Monitoring, Ambulatory , Child, Preschool , Follow-Up Studies , Glomerular Filtration Rate , Humans , Hypertension/etiology , Kidney , Living Donors , Nephrectomy
13.
J Cell Physiol ; 236(5): 4036-4049, 2021 05.
Article in English | MEDLINE | ID: mdl-33151557

ABSTRACT

Percutaneous transluminal renal angioplasty (PTRA) has been used to treat renovascular disease (RVD), a chronic condition characterized by renal ischemia and metabolic abnormalities. Mitochondrial injury has been implicated as a central pathogenic mechanism in RVD, but whether it can be reversed by PTRA remains uncertain. We hypothesized that PTRA attenuates mitochondrial damage, renal injury, and dysfunction in pigs with coexisting renal artery stenosis (RAS) and metabolic syndrome (MetS). Four groups of pigs (n = 6 each) were studied after 16 weeks of diet-induced MetS and RAS (MetS + RAS), MetS + RAS treated 4 weeks earlier with PTRA, and Lean and MetS Sham controls. Single-kidney renal blood flow (RBF) and glomerular filtration rate (GFR) were assessed in vivo with multidetector computed tomography, and renal tubular mitochondrial structure and function and renal injury ex vivo. PTRA successfully restored renal artery patency, but mean arterial pressure remained unchanged. Stenotic kidney RBF and GFR, which fell in MetS + RAS compared to MetS, rose after PTRA. PTRA attenuated MetS + RAS-induced mitochondrial structural abnormalities in tubular cells and peritubular capillary endothelial cells, decreased mitochondrial H2 02 production, and increased renal cytochrome-c oxidase-IV activity and ATP production. PTRA also improved cortical microvascular and peritubular capillary density and ameliorated tubular injury and tubulointerstitial fibrosis in the poststenotic kidney. Importantly, renal mitochondrial damage correlated with poststenotic injury and dysfunction. Renal revascularization attenuated mitochondrial injury and improved renal hemodynamics and function in swine poststenotic kidneys. This study suggests a novel mechanism by which PTRA might be relatively effective in ameliorating mitochondrial damage and improving renal function in coexisting MetS and RAS.


Subject(s)
Angioplasty , Kidney/surgery , Metabolic Syndrome/complications , Metabolic Syndrome/surgery , Mitochondria/pathology , Renal Artery Obstruction/complications , Renal Artery Obstruction/surgery , Animals , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Fibrosis , Hemodynamics , Hypertension/complications , Hypertension/physiopathology , Kidney/blood supply , Kidney/pathology , Kidney/physiopathology , Metabolic Syndrome/physiopathology , Mitochondria/ultrastructure , Oxidative Stress , Renal Artery Obstruction/physiopathology , Swine
14.
J Cell Physiol ; 236(2): 1332-1344, 2021 02.
Article in English | MEDLINE | ID: mdl-32657444

ABSTRACT

Cell stress may give rise to insuperable growth arrest, which is defined as cellular senescence. Stenotic kidney (STK) ischemia and injury induced by renal artery stenosis (RAS) may be associated with cellular senescence. Mesenchymal stem cells (MSCs) decrease some forms of STK injury, but their ability to reverse senescence in RAS remains unknown. We hypothesized that RAS evokes STK senescence, which would be ameliorated by MSCs. Mice were studied after 4 weeks of RAS, RAS treated with adipose tissue-derived MSCs 2 weeks earlier, or sham. STK senescence-associated ß-galactosidase (SA-ß-Gal) activity was measured. Protein and gene expression was used to assess senescence and the senescence-associated secretory phenotype (SASP), and staining for renal fibrosis, inflammation, and capillary density. In addition, senescence was assessed as p16+ and p21+ urinary exosomes in patients with renovascular hypertension (RVH) without or 3 months after autologous adipose tissue-derived MSC delivery, and in healthy volunteers (HV). In RAS mice, STK SA-ß-Gal activity increased, and senescence and SASP marker expression was markedly elevated. MSCs improved renal function, fibrosis, inflammation, and capillary density, and attenuated SA-ß-Gal activity, but most senescence and SASP levels remained unchanged. Congruently, in human RVH, p21+ urinary exosomes were elevated compared to HV, and only slightly improved by MSC, whereas p16+ exosomes remained unchanged. Therefore, RAS triggers renal senescence in both mice and human subjects. MSCs decrease renal injury, but only partly mitigate renal senescence. These observations support exploration of targeted senolytic therapy in RAS.


Subject(s)
Cellular Senescence/genetics , Mesenchymal Stem Cell Transplantation , Renal Artery Obstruction/therapy , beta-Galactosidase/genetics , Adipose Tissue/cytology , Animals , Disease Models, Animal , Exosomes/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/therapy , Kidney/metabolism , Kidney/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Renal Artery Obstruction/genetics , Renal Artery Obstruction/pathology
15.
Sci Rep ; 10(1): 16300, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004888

ABSTRACT

Tissue fibrosis is an important index of renal disease progression. Diffusion-weighted magnetic resonance imaging's (DWI-MRI) apparent diffusion coefficient (ADC) reveals water diffusion is unobstructed by microstructural alterations like fibrosis. We hypothesized that ADC may indicate renal injury and response to therapy in patients with renovascular disease (RVD). RVD patients were treated with medical therapy (MT) and percutaneous transluminal renal angioplasty (MT + PTRA) (n = 11, 3 bilaterally, n = 14 kidneys) or MT (n = 9). ADC and renal hypoxia (R2*) by blood-oxygen-level-dependent MRI were studied before (n = 27) and 3 months after (n = 20) treatment. Twelve patients underwent renal biopsies. Baseline ADC values were correlated with changes in eGFR, serum creatinine (SCr), systolic blood pressure (SBP), renal hypoxia, and renal vein levels of pro-inflammatory marker tumor necrosis-factor (TNF)-α. Renal oxygenation, eGFR, and SCr improved after MT + PTRA. ADC inversely correlated with the histological degree of renal fibrosis, but remained unchanged after MT or MT + PTRA. Basal ADC values correlated modestly with change in SBP, but not in renal hypoxia, TNF-α levels, or renal function. Lower ADC potentially reflects renal injury in RVD patients, but does not change in response to medical or interventional therapy over 3 months. Future studies need to pinpoint indices of kidney recovery potential.


Subject(s)
Kidney/pathology , Renal Artery Obstruction/pathology , Aged , Angioplasty , Biopsy , Diffusion Magnetic Resonance Imaging , Female , Fibrosis , Humans , Kidney/diagnostic imaging , Male , Renal Artery Obstruction/diagnostic imaging , Renal Artery Obstruction/therapy , Treatment Outcome
16.
Curr Opin Cardiol ; 35(6): 627-635, 2020 11.
Article in English | MEDLINE | ID: mdl-32852347

ABSTRACT

PURPOSE OF REVIEW: Renovascular occlusive disease remains a common cause of resistant and rapidly progressive hypertension. The present review summarizes current practice regarding management of renovascular hypertension (RVH). RECENT FINDINGS: Current data using blood oxygen level dependent MR emphasize the tolerance of the kidney to moderate reductions in blood flow and the efficacy of antihypertensive drug therapy for many individuals. Prospective trials have failed to identify benefits of revascularization for moderate disease, either regarding blood pressure or renal function. Antihypertensive drug therapy including renin-angiotensin system blockade is central to management of RVH. Recent and ongoing observational studies report important improvements after revascularization regarding blood pressure, management of refractory or 'flash' pulmonary edema, and survival in specific 'high risk' clinical populations not included in randomized trials. Research directions underscore the role of adjunctive measures, including mitochondrial protection, therapeutic angiogenesis, and cell-based regenerative repair to protect kidney function in RVH. SUMMARY: Clinicians should recognize the potential for disease progression to threaten renal function with severe and prolonged renal ischemia. Improved patient selection for true resistant hypertension with RVH and 'high-risk' clinical manifestations is critical to identify those likely to benefit from renal revascularization.


Subject(s)
Hypertension, Renovascular , Hypertension , Blood Pressure , Humans , Hypertension, Renovascular/diagnosis , Hypertension, Renovascular/etiology , Hypertension, Renovascular/therapy , Prospective Studies , Vascular Surgical Procedures
17.
Hypertension ; 76(2): 497-505, 2020 08.
Article in English | MEDLINE | ID: mdl-32507040

ABSTRACT

The significance of peristenotic collateral circulation (PCC) development around a stenotic renal artery is unknown. We tested the hypothesis that PCC is linked to loss of kidney function and recovery potential in patients with atherosclerotic renovascular disease (ARVD). Thirty-four patients with ARVD were assigned to medical-therapy with or without revascularization based on clinical indications. The PCC was visualized using multidetector computed tomography and defined relative to segmental arteries in patients with essential hypertension. PCC number before and 3 months after treatment was correlated with various renal parameters. Thirty-four stenotic kidneys from 30 patients were analyzed. PCC number correlated inversely with kidney volume. ARVD-stenotic kidneys with baseline PCC (collateral ARVD [C-ARVD], n=13) associated with elevated 24-hour urine protein and stenotic kidney vein level of tumor necrosis factor-α, lower single-kidney volume and blood flow, and greater hypoxia than in stenotic kidneys with no PCC (no collateral ARVD [NC-ARVD], n=17). Revascularization (but not medical-therapy alone) improved stenotic kidney function and reduced inflammation in both NC-ARVD and C-ARVD. In C-ARVD, revascularization also increased stenotic kidney volume, blood flow, and oxygenation to levels comparable to NC-ARVD, and induced PCC regression. However, revascularization improved systolic blood pressure, plasma renin activity, and filtration fraction only in NC-ARVD. Therefore, patients with C-ARVD have greater kidney dysfunction, atrophy, hypoxia, and inflammation compared with patients with NC-ARVD, suggesting that PCC does not effectively protect the stenotic kidney in ARVD. Renal artery revascularization improved in C-ARVD stenotic kidney function, but not hypertension or renin-angiotensin system activation. These observations may help direct management of patients with ARVD.


Subject(s)
Atherosclerosis/physiopathology , Collateral Circulation/physiology , Kidney Diseases/physiopathology , Kidney/physiopathology , Renal Artery Obstruction/physiopathology , Renal Circulation/physiology , Aged , Atherosclerosis/diagnostic imaging , Blood Pressure/physiology , Female , Glomerular Filtration Rate/physiology , Humans , Kidney/diagnostic imaging , Kidney Diseases/diagnostic imaging , Magnetic Resonance Imaging , Male , Middle Aged , Multidetector Computed Tomography , Renal Artery Obstruction/diagnostic imaging
19.
Kidney Int ; 97(4): 793-804, 2020 04.
Article in English | MEDLINE | ID: mdl-32093917

ABSTRACT

Atherosclerotic renovascular disease (ARVD) reduces tissue perfusion and eventually leads to loss of kidney function with limited therapeutic options. Here we describe results of Phase 1a escalating dose clinical trial of autologous mesenchymal stem cell infusion for ARVD. Thirty-nine patients with ARVD were studied on two occasions separated by three months. Autologous adipose-derived mesenchymal stem cells were infused through the renal artery in 21 patients at three different dose levels (1, 2.5 and 5.0 × 105 cells/kg) in seven patients each. We measured renal blood flow, glomerular filtration rate (GFR) (iothalamate and estimated GFR), renal vein cytokine levels, blood pressure, and tissue oxygenation before and three months after stem cell delivery. These indices were compared to those of 18 patients with ARVD matched for age, kidney function and blood pressure receiving medical therapy alone that underwent an identical study protocol. Cultured mesenchymal stem cells were also studied in vitro. For the entire stem cell treated-cohort, mean renal blood flow in the treated stenotic kidney significantly increased after stem cell infusion from (164 to 190 ml/min). Hypoxia, renal vein inflammatory cytokines, and angiogenic biomarkers significantly decreased following stem cell infusion. Mean systolic blood pressure significantly fell (144 to 136 mmHg) and the mean two-kidney GFR (Iothalamate) modestly but significantly increased from (53 to 56 ml/min). Changes in GFR and blood pressure were largest in the high dose stem cell treated individuals. No such changes were observed in the cohort receiving medical treatment alone. Thus, our data demonstrate the potential for autologous mesenchymal stem cell to increase blood flow, GFR and attenuate inflammatory injury in post-stenotic kidneys. The observation that some effects are dose-dependent and related to in-vitro properties of mesenchymal stem cell may direct efforts to maximize potential therapeutic efficacy.


Subject(s)
Mesenchymal Stem Cells , Renal Artery Obstruction , Biomarkers , Blood Pressure , Glomerular Filtration Rate , Humans , Kidney , Renal Artery Obstruction/therapy , Renal Circulation
SELECTION OF CITATIONS
SEARCH DETAIL
...