Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Mol Life Sci ; 80(9): 265, 2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37615749

ABSTRACT

Transient receptor potential cation channel-6 (TRPC6) gene mutations cause familial focal segmental glomerulosclerosis (FSGS), which is inherited as an autosomal dominant disease. In patients with TRPC6-related FSGS, all mutations map to the N- or C-terminal TRPC6 protein domains. Thus far, the majority of TRPC6 mutations are missense resulting in increased or decreased calcium influx; however, the fundamental molecular mechanisms causing cell injury and kidney pathology are unclear. We report a novel heterozygous TRPC6 mutation (V691Kfs*) in a large kindred with no signs of FSGS despite a largely truncated TRPC6 protein. We studied the molecular effects of V691Kfs* TRPC6 mutant using the tridimensional cryo-EM structure of the tetrameric TRPC6 protein. The results indicated that V691 is localized at the pore-forming transmembrane region affecting the ion conduction pathway, and predicted that V691Kfs* causes closure of the ion-conducting pathway leading to channel inactivation. We assessed the impact of V691Kfs* and two previously reported TRPC6 disease mutants (P112Q and G757D) on calcium influx in cells. Our data show that the V691Kfs* fully inactivated the TRCP6 channel-specific calcium influx consistent with a complete loss-of-function phenotype. Furthermore, the V691Kfs* truncation exerted a dominant negative effect on the full-length TRPC6 proteins. In conclusion, the V691Kfs* non-functional truncated TRPC6 is not sufficient to cause FSGS. Our data corroborate recently characterized TRPC6 loss-of-function and gain-of-function mutants suggesting that one defective TRPC6 gene copy is not sufficient to cause FSGS. We underscore the importance of increased rather than reduced calcium influx through TRPC6 for podocyte cell death.


Subject(s)
Glomerulosclerosis, Focal Segmental , Humans , Glomerulosclerosis, Focal Segmental/genetics , TRPC6 Cation Channel/genetics , Calcium , Loss of Function Mutation , Mutation/genetics
2.
BMC Geriatr ; 24(1): 498, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844878

ABSTRACT

BACKGROUND: More than one in two older people wake up several times a night to urinate. Far from being a minor inconvenience, nocturia is associated with poor health outcomes. Given the importance of sleep as a foundation for resilience and healthy ageing, nocturia may promote the development of frailty, which is inextricably linked to physical decline, disability, and mortality. The aim of this scoping review was to collate published evidence on the relationship between nocturia and frailty, using the methodological framework of Arksey and O'Malley, together with the Joanna Briggs Institute methodology as guidance (OSF registration: osf.io/d5ct7). METHODS: Relevant publications were retrieved via PubMed, Embase, the Cochrane Library and Google Scholar. The Rayyan tool was used to facilitate the screening process. Data were extracted by two independent reviewers. 250 publications were initially identified, of which 87 met the eligibility criteria. RESULTS: Most of the evidence came from cross-sectional studies, most of which had been published within the last 5 years. The researchers were diverse, with 27% having a geriatric background. Only few publications established a clear association between nocturia and frailty. Other topics included: the association between nocturia and poor sleep quality and duration; the association between sleep and frailty; the association between frailty, multimorbidity, and age-related changes in the lower urinary tract. CONCLUSION: The findings emphasize the increasing interest and interdisciplinary nature of research into the relationship between frailty, nocturia, lower urinary tract symptoms, and sleep disturbances. Further research is required to enhance understanding, establish causality, and identify potential therapeutic approaches.


Subject(s)
Frailty , Nocturia , Humans , Nocturia/epidemiology , Nocturia/diagnosis , Aged , Frailty/epidemiology , Frailty/diagnosis , Frailty/complications , Frail Elderly , Aged, 80 and over
3.
Nature ; 551(7682): 585-589, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29143823

ABSTRACT

A Western lifestyle with high salt consumption can lead to hypertension and cardiovascular disease. High salt may additionally drive autoimmunity by inducing T helper 17 (TH17) cells, which can also contribute to hypertension. Induction of TH17 cells depends on gut microbiota; however, the effect of salt on the gut microbiome is unknown. Here we show that high salt intake affects the gut microbiome in mice, particularly by depleting Lactobacillus murinus. Consequently, treatment of mice with L. murinus prevented salt-induced aggravation of actively induced experimental autoimmune encephalomyelitis and salt-sensitive hypertension by modulating TH17 cells. In line with these findings, a moderate high-salt challenge in a pilot study in humans reduced intestinal survival of Lactobacillus spp., increased TH17 cells and increased blood pressure. Our results connect high salt intake to the gut-immune axis and highlight the gut microbiome as a potential therapeutic target to counteract salt-sensitive conditions.


Subject(s)
Gastrointestinal Microbiome/drug effects , Lactobacillus/drug effects , Lactobacillus/isolation & purification , Sodium Chloride/pharmacology , Th17 Cells/drug effects , Th17 Cells/immunology , Animals , Autoimmunity/drug effects , Blood Pressure/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/microbiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Feces/microbiology , Humans , Hypertension/chemically induced , Indoleacetic Acids/metabolism , Indoles/metabolism , Intestines/cytology , Intestines/drug effects , Intestines/immunology , Intestines/microbiology , Lactobacillus/immunology , Lymphocyte Activation/drug effects , Lymphocyte Count , Male , Mice , Pilot Projects , Sodium Chloride/administration & dosage , Symbiosis , Th17 Cells/cytology , Tryptophan/metabolism
4.
Lipids Health Dis ; 22(1): 138, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37644527

ABSTRACT

BACKGROUND: Oxylipins, the oxidative metabolites of polyunsaturated fatty acids (PUFAs), serve as key mediators of oxidative stress, inflammatory responses, and vasoactive reactions in vivo. Our previous work has established that hemodialysis affects both long chain fatty acids (LCFAs) and oxylipins in plasma and erythrocytes to varying degrees, which may be responsible for excess cardiovascular complications in end-stage renal disease. In this study, we aimed to determine changes in blood oxylipins during cardiopulmonary bypass (CPB) in patients undergoing cardiac surgery to identify novel biomarkers and potential metabolites of CPB-related complications. We tested the hypothesis that CPB would differentially affect plasma oxylipins and erythrocytes oxylipins. METHODS: We conducted a prospective observational study of 12 patients undergoing elective cardiac surgery with expected CPB procedure. We collected venous and arterial blood samples before CPB, 15 and 45 min after the start of CPB, and 60 min after the end of CPB, respectively. Oxylipins profiling in plasma and erythrocytes was achieved using targeted HPLC-MS mass spectrometry. RESULTS: Our results revealed that most venous plasma diols and hydroxy- oxylipins decreased after CPB initiation, with a continuous decline until the termination of CPB. Nevertheless, no statistically significant alterations were detected in erythrocytes oxylipins at all time points. CONCLUSIONS: CPB decreases numerous diols and hydroxy oxylipins in blood plasma, whereas no changes in erythrocytes oxylipins are observed during this procedure in patients undergoing cardiac surgery. As lipid mediators primarily responsive to CPB, plasma diols and hydroxy oxylipins may serve as potential key biomarkers for CPB-related complications.


Subject(s)
Cardiopulmonary Bypass , Oxylipins , Humans , Cardiopulmonary Bypass/adverse effects , Plasma , Erythrocytes , Fatty Acids
5.
Nephrol Dial Transplant ; 37(12): 2496-2504, 2022 11 23.
Article in English | MEDLINE | ID: mdl-35022790

ABSTRACT

BACKGROUND: Angiotensin-converting enzyme inhibitors (ACEis) have evolved as a first-line therapy for delaying end-stage renal failure (ESRF) in Alport syndrome (AS). The present study tested the hypothesis of a superior nephroprotective potential of an early ACEi intervention, examining a cohort with the COL4A5 missense variant p.(Gly624Asp). METHODS: In this observational cohort study (NCT02378805), 114 individuals with the identical gene variant were explored for age at ESRF and life expectancy in correlation with treatment as endpoints. RESULTS: All 13 untreated hemizygous patients developed ESRF (mean age 48.9 ± 13.7 years), as did 3 very late treated hemizygotes (51.7 ± 4.2 years), with a mean life expectancy of 59.2 ± 9.6 years. All 28 earlier-treated [estimated glomerular filtration rate (eGFR) ≥60 mL/min/1.73 m2] hemizygous patients were still alive and still had not reached ESRF. Therapy minimized the annual loss of their GFR, similar to the annual loss in healthy individuals. Of 65 heterozygotes, 4 untreated individuals developed ESRF at an age of 53.3 ± 20.7 years. None of the treated heterozygous females developed ESRF. CONCLUSIONS: For the first time, this study shows that in AS, early therapy in individuals with missense variants might have the potential to delay renal failure for their lifetime and thus to improve life expectancy and quality of life without the need for renal replacement therapy. Some treated patients have reached their retirement age with still-functioning kidneys, whereas their untreated relatives have reached ESRF at the same or a younger age. Thus, in children with glomerular haematuria, early testing for Alport-related gene variants could lead to timely nephroprotective intervention.


Subject(s)
Kidney Failure, Chronic , Nephritis, Hereditary , Adult , Aged , Female , Humans , Middle Aged , Collagen Type IV/genetics , Heterozygote , Kidney Failure, Chronic/genetics , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/genetics , Prospective Studies , Quality of Life
6.
Int J Mol Sci ; 23(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35743312

ABSTRACT

Metabolic syndrome is a significant worldwide public health challenge and is inextricably linked to adverse renal and cardiovascular outcomes. The inhibition of the transient receptor potential cation channel subfamily C member 6 (TRPC6) has been found to ameliorate renal outcomes in the unilateral ureteral obstruction (UUO) of accelerated renal fibrosis. Therefore, the pharmacological inhibition of TPRC6 could be a promising therapeutic intervention in the progressive tubulo-interstitial fibrosis in hypertension and metabolic syndrome. In the present study, we hypothesized that the novel selective TRPC6 inhibitor SH045 (larixyl N-methylcarbamate) ameliorates UUO-accelerated renal fibrosis in a New Zealand obese (NZO) mouse model, which is a polygenic model of metabolic syndrome. The in vivo inhibition of TRPC6 by SH045 markedly decreased the mRNA expression of pro-fibrotic markers (Col1α1, Col3α1, Col4α1, Acta2, Ccn2, Fn1) and chemokines (Cxcl1, Ccl5, Ccr2) in UUO kidneys of NZO mice compared to kidneys of vehicle-treated animals. Renal expressions of intercellular adhesion molecule 1 (ICAM-1) and α-smooth muscle actin (α-SMA) were diminished in SH045- versus vehicle-treated UUO mice. Furthermore, renal inflammatory cell infiltration (F4/80+ and CD4+) and tubulointerstitial fibrosis (Sirius red and fibronectin staining) were ameliorated in SH045-treated NZO mice. We conclude that the pharmacological inhibition of TRPC6 might be a promising antifibrotic therapeutic method to treat progressive tubulo-interstitial fibrosis in hypertension and metabolic syndrome.


Subject(s)
Hypertension , Kidney Diseases , Metabolic Syndrome , Ureteral Obstruction , Animals , Disease Models, Animal , Fibrosis , Hypertension/metabolism , Kidney/metabolism , Kidney Diseases/etiology , Kidney Diseases/genetics , Metabolic Syndrome/complications , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Mice , Mice, Obese , New Zealand , Obesity/complications , Obesity/drug therapy , Obesity/metabolism , TRPC6 Cation Channel/metabolism , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy , Ureteral Obstruction/genetics
7.
Circulation ; 142(2): 133-149, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32524868

ABSTRACT

BACKGROUND: High blood pressure is the primary risk factor for cardiovascular death worldwide. Autosomal dominant hypertension with brachydactyly clinically resembles salt-resistant essential hypertension and causes death by stroke before 50 years of age. We recently implicated the gene encoding phosphodiesterase 3A (PDE3A); however, in vivo modeling of the genetic defect and thus showing an involvement of mutant PDE3A is lacking. METHODS: We used genetic mapping, sequencing, transgenic technology, CRISPR-Cas9 gene editing, immunoblotting, and fluorescence resonance energy transfer. We identified new patients, performed extensive animal phenotyping, and explored new signaling pathways. RESULTS: We describe a novel mutation within a 15 base pair (bp) region of the PDE3A gene and define this segment as a mutational hotspot in hypertension with brachydactyly. The mutations cause an increase in enzyme activity. A CRISPR/Cas9-generated rat model, with a 9-bp deletion within the hotspot analogous to a human deletion, recapitulates hypertension with brachydactyly. In mice, mutant transgenic PDE3A overexpression in smooth muscle cells confirmed that mutant PDE3A causes hypertension. The mutant PDE3A enzymes display consistent changes in their phosphorylation and an increased interaction with the 14-3-3θ adaptor protein. This aberrant signaling is associated with an increase in vascular smooth muscle cell proliferation and changes in vessel morphology and function. CONCLUSIONS: The mutated PDE3A gene drives mechanisms that increase peripheral vascular resistance causing hypertension. We present 2 new animal models that will serve to elucidate the underlying mechanisms further. Our findings could facilitate the search for new antihypertensive treatments.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Hypertension/genetics , Mutation , Alleles , Amino Acid Substitution , Animals , Animals, Genetically Modified , Arterial Pressure , Biomarkers/blood , Biomarkers/urine , Brachydactyly/diagnosis , Brachydactyly/genetics , CRISPR-Cas Systems , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , DNA Mutational Analysis , Disease Models, Animal , Enzyme Activation , Gene Targeting , Genetic Association Studies/methods , Genotype , Immunohistochemistry , Isoenzymes , Male , Pedigree , Phenotype , Radiography , Rats , Renin-Angiotensin System/genetics
8.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768842

ABSTRACT

Endothelial dysfunction (ED) comes with age, even without overt vessel damage such as that which occurs in atherosclerosis and diabetic vasculopathy. We hypothesized that aging would affect the downstream signalling of the endothelial nitric oxide (NO) system in the vascular smooth muscle (VSM). With this in mind, resistance mesenteric arteries were isolated from 13-week (juvenile) and 40-week-old (aged) mice and tested under isometric conditions using wire myography. Acetylcholine (ACh)-induced relaxation was reduced in aged as compared to juvenile vessels. Pretreatment with L-NAME, which inhibits nitrix oxide synthases (NOS), decreased ACh-mediated vasorelaxation, whereby differences in vasorelaxation between groups disappeared. Endothelium-independent vasorelaxation by the NO donor sodium nitroprusside (SNP) was similar in both groups; however, SNP bolus application (10-6 mol L-1) as well as soluble guanylyl cyclase (sGC) activation by runcaciguat (10-6 mol L-1) caused faster responses in juvenile vessels. This was accompanied by higher cGMP concentrations and a stronger response to the PDE5 inhibitor sildenafil in juvenile vessels. Mesenteric arteries and aortas did not reveal apparent histological differences between groups (van Gieson staining). The mRNA expression of the α1 and α2 subunits of sGC was lower in aged animals, as was PDE5 mRNA expression. In conclusion, vasorelaxation is compromised at an early age in mice even in the absence of histopathological alterations. Vascular smooth muscle sGC is a key element in aged vessel dysfunction.


Subject(s)
Mesenteric Arteries/physiology , Soluble Guanylyl Cyclase/physiology , Acetylcholine/pharmacology , Age Factors , Animals , Aorta/metabolism , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Guanylate Cyclase/metabolism , Male , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
9.
Circulation ; 139(11): 1407-1421, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30586752

ABSTRACT

BACKGROUND: Arterial hypertension and its organ sequelae show characteristics of T cell-mediated inflammatory diseases. Experimental anti-inflammatory therapies have been shown to ameliorate hypertensive end-organ damage. Recently, the CANTOS study (Canakinumab Antiinflammatory Thrombosis Outcome Study) targeting interleukin-1ß demonstrated that anti-inflammatory therapy reduces cardiovascular risk. The gut microbiome plays a pivotal role in immune homeostasis and cardiovascular health. Short-chain fatty acids (SCFAs) are produced from dietary fiber by gut bacteria and affect host immune homeostasis. Here, we investigated effects of the SCFA propionate in 2 different mouse models of hypertensive cardiovascular damage. METHODS: To investigate the effect of SCFAs on hypertensive cardiac damage and atherosclerosis, wild-type NMRI or apolipoprotein E knockout-deficient mice received propionate (200 mmol/L) or control in the drinking water. To induce hypertension, wild-type NMRI mice were infused with angiotensin II (1.44 mg·kg-1·d-1 subcutaneous) for 14 days. To accelerate the development of atherosclerosis, apolipoprotein E knockout mice were infused with angiotensin II (0.72 mg·kg-1·d-1 subcutaneous) for 28 days. Cardiac damage and atherosclerosis were assessed using histology, echocardiography, in vivo electrophysiology, immunofluorescence, and flow cytometry. Blood pressure was measured by radiotelemetry. Regulatory T cell depletion using PC61 antibody was used to examine the mode of action of propionate. RESULTS: Propionate significantly attenuated cardiac hypertrophy, fibrosis, vascular dysfunction, and hypertension in both models. Susceptibility to cardiac ventricular arrhythmias was significantly reduced in propionate-treated angiotensin II-infused wild-type NMRI mice. Aortic atherosclerotic lesion area was significantly decreased in propionate-treated apolipoprotein E knockout-deficient mice. Systemic inflammation was mitigated by propionate treatment, quantified as a reduction in splenic effector memory T cell frequencies and splenic T helper 17 cells in both models, and a decrease in local cardiac immune cell infiltration in wild-type NMRI mice. Cardioprotective effects of propionate were abrogated in regulatory T cell-depleted angiotensin II-infused mice, suggesting the effect is regulatory T cell-dependent. CONCLUSIONS: Our data emphasize an immune-modulatory role of SCFAs and their importance for cardiovascular health. The data suggest that lifestyle modifications leading to augmented SCFA production could be a beneficial nonpharmacological preventive strategy for patients with hypertensive cardiovascular disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aortic Diseases/drug therapy , Arrhythmias, Cardiac/prevention & control , Atherosclerosis/drug therapy , Cardiomegaly/prevention & control , Hypertension/drug therapy , Propionates/pharmacology , Angiotensin II , Animals , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/physiopathology , Arterial Pressure/drug effects , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cardiomegaly/immunology , Cardiomegaly/physiopathology , Disease Models, Animal , Hypertension/chemically induced , Hypertension/immunology , Hypertension/physiopathology , Male , Mice, Knockout, ApoE , Plaque, Atherosclerotic , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
10.
Annu Rev Pharmacol Toxicol ; 57: 417-436, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27732801

ABSTRACT

Excess visceral adipose tissue is associated with increased risk of high blood pressure, lipid disorders, type 2 diabetes, and cardiovascular disease. Adipose tissue is an endocrine organ with multiple humoral and metabolic roles in regulating whole-body physiology. However, perivascular adipose tissue (PVAT) also plays a functional role in regulating the contractile state of the underlying smooth muscle cell layer. Work during the past decade has shown that this adipose-vascular coupling is achieved by production of numerous substances released from PVAT. Animal disease models have been instrumental in identifying biological and pathophysiological functions of this regulation. These studies have produced strong evidence that alterations in the paracrine control of PVAT in the regulation of arterial tone contribute to vascular dysfunction in obesity, hypertension, and cardiometabolic disease. Perivascular relaxing factors, or perhaps their putative targets, might represent exciting new targets for the prevention and treatment of cardiovascular and metabolic diseases.


Subject(s)
Adipokines/metabolism , Adipose Tissue/metabolism , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Adipose Tissue/drug effects , Animals , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/drug effects , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Muscle, Smooth, Vascular/drug effects , Obesity/drug therapy , Obesity/metabolism
11.
Blood Purif ; 49(3): 356-363, 2020.
Article in English | MEDLINE | ID: mdl-31812967

ABSTRACT

BACKGROUND/AIMS: Trajectory of heart rate variability (HRV) represents a noninvasive real-time measure of autonomous nervous system (ANS) and carries the capability of providing new insights into the hemodynamic compensation reserve during hemodialysis (HD). However, studies on HRV reproducibility during HD are scarce and did not refer to different reading periods. In this observational study, we aimed to establish the best suited and most reliable and reproducible HRV index in routine HD treatments including different reading rates. METHODS: HRV was characterized by standardized mathematical variation expressions of R/R' intervals: SD of all R/R' intervals (ms), square root of the root mean square of the sum of all differences between adjacent R/R' intervals (ms), percentage of consecutive R/R' intervals that differ by >50 ms (%), low-frequency spectral analysis HRV (LF, expressing sympathetic activity), and high-frequency HRV (HF, expressing parasympathetic activity). To compare robustness of these HRV indices during HD procedures, we compared HRV indices means between different HD sessions and controlled for association with clinical parameters. RESULTS: In 72 HD treatments of 34 patients, we detected the highest reproducibility (89%) of HRV measures when analyzing the low-frequency to high-frequency (LF/HF) ratio in long-term (3 h) readings. Long-term LF/HF was able to discriminate -between patients with and without heart failure NYHA classes ≥3 (p = 0.009) and type 2 diabetes (p = 0.023). We were unable to study relationships between ANS and intradialytic complications because they did not appear in our cohort. Short-term readings of HRV indices did not show any significance of pattern change during HD. CONCLUSION: In summary, our data provide evidence for high robustness of long-term LF/HF in analyzing HRV in HD patients using future automated monitoring systems. For short-term analysis, mathematical real-time analysis must evolve.


Subject(s)
Heart Rate , Renal Dialysis , Adult , Aged , Diabetes Mellitus, Type 2/diagnosis , Female , Heart Failure/diagnosis , Humans , Kidney Failure, Chronic/therapy , Male , Middle Aged , Reproducibility of Results
12.
Cell Physiol Biochem ; 52(6): 1484-1502, 2019.
Article in English | MEDLINE | ID: mdl-31099508

ABSTRACT

BACKGROUND/AIMS: The transient receptor potential cation channel subfamily C member 6 (TRPC6) is a Ca2+-permeable nonselective cation channel and has received recent attention because of its capability to promote chronic kidney disease (CKD). The aims of this study were (i) to examine whether deletion of TRPC6 impacts on renal fibrosis and inflammatory cell infiltration in an early CKD model of unilateral ureter obstruction (UUO) in mice; and (ii) whether TRPC6-deficiency as well as UUO affect the regulation of TRPC expression in murine kidneys. METHODS: Wild-type (WT), Trpc6-knockout (Trpc6-/-) and New Zealand obese (NZO) mice underwent sham operation or unilateral ureteral obstruction (UUO). The kidneys were harvested 7 days after surgery. We examined renal fibrosis and inflammatory cell infiltration by histological and immunohistochemical staining. The mRNA expression of TRPC members and markers of fibrosis and inflammation in kidney were assessed by using real-time quantitative reverse transcription PCR. RESULTS: Histological and immunohistochemical analyses revealed less inflammatory cell infiltration (F4/80 and CD3) in UUO kidneys of Trpc6-/- mice compared to UUO kidneys of WT mice as well as less fibrosis. Genomic deletion of TRPC6 also affected the expression of pro-fibrotic genes in UUO Trpc6-/- kidneys compared to UUO WT kidneys while the expression of pro-inflammatory genes did not differ. UUO caused marked up-regulation of Trpc6 and down-regulation of Trpc1 mRNA in kidneys of WT and NZO mice. Trpc3 mRNA expression was significantly elevated in kidneys of Trpc6-/- mice underwent UUO while the levels did not change in kidneys of neither WT nor in NZO mice underwent UUO. CONCLUSION: TRPC6 contributes to renal fibrosis and immune cell infiltration in the UUO mouse model. Therefore, inhibition of TRPC6 emerges as a promising novel therapeutic strategy for treatment of chronic kidney failure in chronic obstructive nephropathy. However, confounding genomic and non-genomic effects of other TRPC channels should be taken into consideration to fully comprehend the renoprotective potential of targeting TRPC6 therapeutically under chronic kidney damaging conditions.


Subject(s)
Gene Expression Regulation , Kidney/pathology , TRPC Cation Channels/genetics , Ureteral Obstruction/genetics , Animals , Disease Models, Animal , Fibrosis , Gene Deletion , Inflammation/complications , Inflammation/genetics , Inflammation/pathology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , RNA, Messenger/analysis , RNA, Messenger/genetics , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , TRPC6 Cation Channel , Up-Regulation , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
13.
Arterioscler Thromb Vasc Biol ; 38(10): 2371-2381, 2018 10.
Article in English | MEDLINE | ID: mdl-30354206

ABSTRACT

Objective- This study examined whether caveolae position CaV3.2 (T-type Ca2+ channel encoded by the α-3.2 subunit) sufficiently close to RyR (ryanodine receptors) for extracellular Ca2+ influx to trigger Ca2+ sparks and large-conductance Ca2+-activated K+ channel feedback. Approach and Results- Using smooth muscle cells from mouse mesenteric arteries, the proximity ligation assay confirmed that CaV3.2 reside within 40 nm of caveolin 1, a key caveolae protein. Methyl-ß-cyclodextrin, a cholesterol depleting agent that disrupts caveolae, suppressed CaV3.2 activity along with large-conductance Ca2+-activated K+-mediated spontaneous transient outward currents in cells from C57BL/6 but not CaV3.2-/- mice. Genetic deletion of caveolin 1, a perturbation that prevents caveolae formation, also impaired spontaneous transient outward current production but did so without impairing Ca2+ channel activity, including CaV3.2. These observations indicate a mistargeting of CaV3.2 in caveolin 1-/- mice, a view supported by a loss of Ni2+-sensitive Ca2+ spark generation and colocalization signal (CaV3.2-RyR) from the proximity ligation assay. Vasomotor and membrane potential measurements confirmed that cellular disruption of the CaV3.2-RyR axis functionally impaired the ability of large-conductance Ca2+-activated K+ to set tone in pressurized caveolin 1-/- arteries. Conclusions- Caveolae play a critical role in protein targeting and preserving the close structural relationship between CaV3.2 and RyR needed to drive negative feedback control in resistance arteries.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium Signaling , Caveolae/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Calcium Channels, T-Type/deficiency , Calcium Channels, T-Type/genetics , Caveolin 1/genetics , Caveolin 1/metabolism , Feedback, Physiological , Male , Membrane Potentials , Mesenteric Arteries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Ryanodine Receptor Calcium Release Channel/metabolism , Vasoconstriction , Vasodilation
14.
J Physiol ; 596(20): 4863-4877, 2018 10.
Article in English | MEDLINE | ID: mdl-30146760

ABSTRACT

KEY POINTS: In arterial smooth muscle, Ca2+ sparks are elementary Ca2+ -release events generated by ryanodine receptors (RyRs) to cause vasodilatation by opening maxi Ca2+ -sensitive K+ (BKCa ) channels. This study elucidated the contribution of T-type Cav 3.2 channels in caveolae and their functional interaction with L-type Cav 1.2 channels to trigger Ca2+ sparks in vascular smooth muscle cells (VSMCs). Our data demonstrate that L-type Cav 1.2 channels provide the predominant Ca2+ pathway for the generation of Ca2+ sparks in murine arterial VSMCs. T-type Cav 3.2 channels represent an additional source for generation of VSMC Ca2+ sparks. They are located in pit structures of caveolae to provide locally restricted, tight coupling between T-type Cav 3.2 channels and RyRs to ignite Ca2+ sparks. ABSTRACT: Recent data suggest that T-type Cav 3.2 channels in arterial vascular smooth muscle cells (VSMCs) and pits structure of caveolae could contribute to elementary Ca2+ signalling (Ca2+ sparks) via ryanodine receptors (RyRs) to cause vasodilatation. While plausible, their precise involvement in igniting Ca2+ sparks remains largely unexplored. The goal of this study was to elucidate the contribution of caveolar Cav 3.2 channels and their functional interaction with Cav 1.2 channels to trigger Ca2+ sparks in VSMCs from mesenteric, tibial and cerebral arteries. We used tamoxifen-inducible smooth muscle-specific Cav 1.2-/- (SMAKO) mice and laser scanning confocal microscopy to assess Ca2+ spark generation in VSMCs. Ni2+ , Cd2+ and methyl-ß-cyclodextrin were used to inhibit Cav 3.2 channels, Cav 1.2 channels and caveolae, respectively. Ni2+ (50 µmol L-1 ) and methyl-ß-cyclodextrin (10 mmol L-1 ) decreased Ca2+ spark frequency by ∼20-30% in mesenteric VSMCs in a non-additive manner, but failed to inhibit Ca2+ sparks in tibial and cerebral artery VSMCs. Cd2+ (200 µmol L-1 ) suppressed Ca2+ sparks in mesenteric arteries by ∼70-80%. A similar suppression of Ca2+ sparks was seen in mesenteric artery VSMCs of SMAKO mice. The remaining Ca2+ sparks were fully abolished by Ni2+ or methyl-ß-cyclodextrin. Our data demonstrate that Ca2+ influx through CaV 1.2 channels is the primary means of triggering Ca2+ sparks in murine arterial VSMCs. CaV 3.2 channels, localized to caveolae and tightly coupled to RyR, provide an additional Ca2+ source for Ca2+ spark generation in mesenteric, but not tibial and cerebral, arteries.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Calcium Signaling , Mesenteric Arteries/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Caveolae/metabolism , Cells, Cultured , Male , Mesenteric Arteries/cytology , Mice , Myocytes, Smooth Muscle/metabolism
15.
Microcirculation ; 25(1)2018 01.
Article in English | MEDLINE | ID: mdl-29211322

ABSTRACT

Resistant hypertension is defined as high blood pressure that remains uncontrolled despite treatment with at least three antihypertensive drugs at adequate doses. Resistant hypertension is an increasingly common clinical problem in older age, obesity, diabetes, sleep apnea, and chronic kidney disease. Although the direct vasodilator minoxidil was introduced in the early 1970s, only recently has this drug been shown to be particularly effective in a subgroup of patients with treatment-resistant or uncontrolled hypertension. This pharmacological approach is interesting from a mechanistic perspective as minoxidil is the only clinically used K+ channel opener today, which targets a subclass of K+ channels, namely KATP channels in VSMCs. Beside KATP channels, two other classes of VSMC K+ channels could represent novel effective targets for treatment of resistant hypertension, namely Kv 7 (KCNQ) and inward rectifier potassium (Kir 2.1) channels. Interestingly, these channels are unique among VSMC potassium channels. First, both have been implicated in the control of microvascular tone by perivascular adipose tissue. Second, they exhibit biophysical properties strongly controlled and regulated by membrane voltage, but not intracellular calcium. This review focuses on Kv 7 (Kv 7.1-5) and Kir (Kir 2.1) channels in VSMCs as potential novel drug targets for treatment of resistant hypertension, particularly in comorbid conditions such as obesity and metabolic syndrome.


Subject(s)
Adipose Tissue/physiology , Potassium Channels, Inwardly Rectifying/physiology , Potassium Channels, Voltage-Gated/physiology , Animals , Humans , Hypertension/drug therapy , Muscle, Smooth, Vascular/cytology
16.
Cardiovasc Drugs Ther ; 32(5): 481-502, 2018 10.
Article in English | MEDLINE | ID: mdl-30171461

ABSTRACT

Perivascular adipose tissue (PVAT) refers to the local aggregate of adipose tissue surrounding the vascular tree, exhibiting phenotypes from white to brown and beige adipocytes. Although PVAT has long been regarded as simply a structural unit providing mechanical support to vasculature, it is now gaining reputation as an integral endocrine/paracrine component, in addition to the well-established modulator endothelium, in regulating vascular tone. Since the discovery of anti-contractile effect of PVAT in 1991, the use of multiple rodent models of reduced amounts of PVAT has revealed its regulatory role in vascular remodeling and cardiovascular implications, including atherosclerosis. PVAT does not only release PVAT-derived relaxing factors (PVRFs) to activate multiple subsets of endothelial and vascular smooth muscle potassium channels and anti-inflammatory signals in the vasculature, but it does also provide an interface for neuron-adipocyte interactions in the vascular wall to regulate arterial vascular tone. In this review, we outline our current understanding towards PVAT and attempt to provide hints about future studies that can sharpen the therapeutic potential of PVAT against cardiovascular diseases and their complications.


Subject(s)
Adipocytes/metabolism , Adipokines/metabolism , Adipose Tissue/metabolism , Blood Vessels/metabolism , Vascular Diseases/metabolism , Adipocytes/pathology , Adipose Tissue/innervation , Adipose Tissue/physiopathology , Adiposity , Animals , Blood Vessels/innervation , Blood Vessels/physiopathology , Cellular Microenvironment , Humans , MicroRNAs/metabolism , Paracrine Communication , Phenotype , Signal Transduction , Vascular Diseases/diagnosis , Vascular Diseases/physiopathology , Vasodilation
17.
Gerontology ; 64(2): 118-126, 2018.
Article in English | MEDLINE | ID: mdl-29183010

ABSTRACT

BACKGROUND: Mild-to-moderate chronic kidney disease (CKD G3a) is prevalent in older adults. Substantial evidence suggests that individuals with advanced CKD face a high risk for common geriatric conditions, like functional impairment and cognitive decline, whereas the relationships between mild-to-moderate CKD and functional impairment and cognitive decline, but also poor nutritional status and mood disorders, are still unclear. OBJECTIVE: The aim of this study was to explore associations between mild-to-moderate CKD and impairments in the core domains of geriatric assessment (GA) in a large cohort of community-dwelling older adults. METHODS: This was a cross-sectional analysis of 1,476 participants of the Berlin Aging Study II. Study participants were stratified as to presence or absence of CKD G3a (estimated glomerular filtration rate [eGFR] 45-59 mL/min/1.73 m2 vs. eGFR ≥60 mL/min/1.73 m2). GA comprised the following instruments: the Activities of Daily Living Scale (ADL), the Timed up and Go (TUG), the Tinetti test (Tinetti), the Mini-Mental-State Examination (MMSE), the Geriatric Depression Scale (GDS), and the Mini Nutritional Assessment (MNA). We used logistic regression models to estimate multivariable-adjusted associations between CKD G3a and impairments in the respective domains. RESULTS: A total of 282 subjects with mild-to-moderate CKD (CKD G3a) were identified (19.1%). Overall, the prevalence of impairments identified was higher among subjects with compared to without CKD G3a (21 vs. 15.9%, p = 0.043). In multivariable-adjusted models, CKD G3a was consistently associated with increased odds of an impaired gait performance as to the TUG (adjusted odds ratio 2.06, 95% CI 1.04-4.09). In contrast, on average, individuals with and without CKD G3a did not differ as to their results in the MMSE, the ADL, the MNA, and the GDS. CONCLUSION: GA identified impairments in 21 versus 15.9% of older adults with and without mild-to-moderate CKD, respectively. However, except for an increased likelihood of impaired gait performance (TUG) with mild-to-moderate CKD, we did not find independent associations between mild-to-moderate CKD and geriatric conditions.


Subject(s)
Renal Insufficiency, Chronic/epidemiology , Activities of Daily Living , Aged , Aging/physiology , Aging/psychology , Berlin/epidemiology , Cognitive Dysfunction/complications , Cross-Sectional Studies , Female , Geriatric Assessment , Glomerular Filtration Rate , Humans , Logistic Models , Male , Multivariate Analysis , Prevalence , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/physiopathology
18.
Gerontology ; 63(3): 201-209, 2017.
Article in English | MEDLINE | ID: mdl-28231583

ABSTRACT

BACKGROUND: In aging populations with an ever-growing burden of risk factors such as obesity, diabetes, and hypertension, chronic kidney disease (CKD) is on the rise. However, little is known about its exact prevalence among elderly adults, and often albuminuria is not included in the definition of CKD. Moreover, novel equations for the estimated glomerular filtration rate (eGFR) have recently emerged, which have not been applied comprehensively to older adults. Data on CKD awareness among the elderly are sparse. OBJECTIVES: To determine the prevalence of CKD among older adults by eGFR and albumin/creatinine ratio (ACR), compare the performance of 6 established and novel eGFR formulas, explore risk factors, and determine the awareness of CKD in a large cohort of community-dwelling elderly from Germany. METHODS: A total of 1,628 subjects from the Berlin Aging Study II (BASE-II) were included in this analysis (mean age 68.7 years; 51.2% female). Extensive cross-sectional data on sociodemographics, lifestyle, medication, and diagnoses were inquired during structured interviews and a medical examination, and blood and urine parameters were measured. RESULTS: In all, 77.1% of the subjects had hypertension, 12.4% had diabetes, and 18.3% were obese. The prevalence of CKD strongly depended on the eGFR equations used: 25.4% (full age spectrum [FAS] equation), 24.6% (Berlin Initiative Study), 23.1% (Lund-Malmö revised), 19.3% (Cockcroft-Gault), 16.4% (Chronic Kidney Disease-Epidemiology Collaboration [CKD-EPI]), and 14.7% (Modification of Diet in Renal Disease [MDRD]). Of the subjects with an eGFRFAS <60 mL/min/1.73 m2 and/or an ACR >30 mg/g, only 3.9% were aware of having CKD. Polypharmacy, age, BMI, coronary artery disease, non-HDL cholesterol, and female sex were independently associated with CKD. CONCLUSIONS: CKD is prevalent among older adults in Germany, but awareness is low. The FAS equation detects higher rates of CKD than MDRD and CKD-EPI, which are most widely used at present. Also, when CKD is defined based on eGFR and albuminuria, considerably more people are identified than by eGFR alone. Finally, polypharmacy is associated with an increased risk for CKD in the elderly.


Subject(s)
Renal Insufficiency, Chronic/epidemiology , Aged , Aged, 80 and over , Aging/physiology , Albuminuria/epidemiology , Berlin/epidemiology , Cross-Sectional Studies , Female , Glomerular Filtration Rate , Humans , Male , Middle Aged , Polypharmacy , Prevalence , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/physiopathology , Risk Factors
19.
J Am Soc Nephrol ; 27(9): 2771-83, 2016 09.
Article in English | MEDLINE | ID: mdl-26892346

ABSTRACT

FSGS is a CKD with heavy proteinuria that eventually progresses to ESRD. Hereditary forms of FSGS have been linked to mutations in the transient receptor potential cation channel, subfamily C, member 6 (TRPC6) gene encoding a nonselective cation channel. Most of these TRPC6 mutations cause a gain-of-function phenotype, leading to calcium-triggered podocyte cell death, but the underlying molecular mechanisms are unclear. We studied the molecular effect of disease-related mutations using tridimensional in silico modeling of tetrameric TRPC6. Our results indicated that G757 is localized in a domain forming a TRPC6-TRPC6 interface and predicted that the amino acid exchange G757D causes local steric hindrance and disruption of the channel complex. Notably, functional characterization of model interface domain mutants suggested a loss-of-function phenotype. We then characterized 19 human FSGS-related TRPC6 mutations, the majority of which caused gain-of-function mutations. However, five mutations (N125S, L395A, G757D, L780P, and R895L) caused a loss-of-function phenotype. Coexpression of wild-type TRPC6 and TRPC6 G757D, mimicking heterozygosity observed in patients, revealed a dominant negative effect of TRPC6 G757D. Our comprehensive analysis of human disease-causing TRPC6 mutations reveals loss of TRPC6 function as an additional concept of hereditary FSGS and provides molecular insights into the mechanism responsible for the loss-of-function phenotype of TRPC6 G757D in humans.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Mutation , TRPC Cation Channels/genetics , DNA Mutational Analysis , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , TRPC6 Cation Channel
20.
Development ; 140(8): 1720-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23533173

ABSTRACT

Arteriogenesis requires growth of pre-existing arteriolar collateral networks and determines clinical outcome in arterial occlusive diseases. Factors responsible for the development of arteriolar collateral networks are poorly understood. The Notch ligand Delta-like 4 (Dll4) promotes arterial differentiation and restricts vessel branching. We hypothesized that Dll4 may act as a genetic determinant of collateral arterial networks and functional recovery in stroke and hind limb ischemia models in mice. Genetic loss- and gain-of-function approaches in mice showed that Dll4-Notch signaling restricts pial collateral artery formation by modulating arterial branching morphogenesis during embryogenesis. Adult Dll4(+/-) mice showed increased pial collateral numbers, but stroke volume upon middle cerebral artery occlusion was not reduced compared with wild-type littermates. Likewise, Dll4(+/-) mice showed reduced blood flow conductance after femoral artery occlusion, and, despite markedly increased angiogenesis, tissue ischemia was more severe. In peripheral arteries, loss of Dll4 adversely affected excitation-contraction coupling in arterial smooth muscle in response to vasopressor agents and arterial vessel wall adaption in response to increases in blood flow, collectively contributing to reduced flow reserve. We conclude that Dll4-Notch signaling modulates native collateral formation by acting on vascular branching morphogenesis during embryogenesis. Dll4 furthermore affects tissue perfusion by acting on arterial function and structure. Loss of Dll4 stimulates collateral formation and angiogenesis, but in the context of ischemic diseases such beneficial effects are overruled by adverse functional changes, demonstrating that ischemic recovery is not solely determined by collateral number but rather by vessel functionality.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Ischemia/physiopathology , Membrane Proteins/metabolism , Microvessels/embryology , Morphogenesis/physiology , Neovascularization, Physiologic/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Analysis of Variance , Animals , Calcium-Binding Proteins , Immunohistochemistry , Ischemia/metabolism , Mice , Microvessels/physiology , Real-Time Polymerase Chain Reaction , Regional Blood Flow/physiology , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL