Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.663
Filter
2.
Diabetes Obes Metab ; 26(7): 2554-2566, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38699780

ABSTRACT

Fibrosis is a common feature of more than 50 different diseases and the cause of more than 35% of deaths worldwide, of which liver, kidney, skin, heart and, recently, lungs are receiving the most attention. Tissue changes, resulting in loss of organ function, are both a cause and consequence of disease and outcome. Fibrosis is caused by an excess deposition of extracellular matrix proteins, which over time results in impaired organ function and organ failure, and the pathways leading to increased fibroblast activation are many. This narrative review investigated the common denominator of fibrosis, fibroblasts, and the activation of fibroblasts, in response to excess energy consumption in liver, kidney, heart, skin and lung fibrosis. Fibroblasts are the main drivers of organ function loss in lung, liver, skin, heart and kidney disease. Fibroblast activation in response to excess energy consumption results in the overproduction of a range of collagens, of which types I, III and VI seem to be the essential drivers of disease progression. Fibroblast activation may be quantified in serum, enabling profiling and selection of patients. Activation of fibroblasts results in the overproduction of collagens, which deteriorates organ function. Patient profiling of fibroblast activities in serum, quantified as collagen production, may identify an organ death trajectory, better enabling identification of the right treatment for use in different metabolic interventions. As metabolically activated patients have highly elevated risk of kidney, liver and heart failure, it is essential to identify which organ to treat first and monitor organ status to correct treatment regimes. In direct alignment with this, it is essential to identify the right patients with the right organ deterioration trajectory for enrolment in clinical studies.


Subject(s)
Fibroblasts , Fibrosis , Metabolic Syndrome , Humans , Fibroblasts/metabolism , Metabolic Syndrome/metabolism , Sclerosis , Kidney Diseases/physiopathology , Collagen/metabolism
4.
Pediatr Blood Cancer ; 71(7): e31002, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38644595

ABSTRACT

BACKGROUND: Tricuspid regurgitation velocity (TRV), measured by echocardiography, is a surrogate marker for pulmonary hypertension. Limited pediatric studies have considered the association between TRV and surrogate markers of end-organ disease. METHODS: We conducted a cross-sectional study that evaluated the prevalence of elevated TRV ≥2.5 m/s and its associations with renal and cerebrovascular outcomes in children with sickle cell disease (SCD) 1-21 years of age in two large sickle cell cohorts, the University of Alabama at Birmingham (UAB) sickle cell cohort, and the Sickle Cell Clinical Research and Intervention Program (SCCRIP) cohort at St. Jude Children's Research Hospital. We hypothesized that patients with SCD and elevated TRV would have higher odds of having either persistent albuminuria or cerebrovascular disease. RESULTS: We identified 166 children from the UAB cohort (mean age: 13.49 ± 4.47 years) and 325 children from the SCCRIP cohort (mean age: 13.41 ± 3.99 years) with echocardiograms. The prevalence of an elevated TRV was 21% in both UAB and SCCRIP cohorts. Elevated TRV was significantly associated with cerebrovascular disease (odds ratio [OR] 1.88, 95% confidence interval [CI]: 1.12-3.15; p = .017) and persistent albuminuria (OR 1.81, 95% CI: 1.07-3.06; p = .028) after adjusting for age, sex, treatment, and site. CONCLUSION: This cross-sectional, multicenter study identifies associations between surrogate markers of pulmonary hypertension with kidney disease and cerebrovascular disease. A prospective study should be performed to evaluate the longitudinal outcomes for patients with multiple surrogate markers of end-organ disease.


Subject(s)
Anemia, Sickle Cell , Cerebrovascular Disorders , Tricuspid Valve Insufficiency , Humans , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/physiopathology , Male , Female , Child , Adolescent , Tricuspid Valve Insufficiency/etiology , Tricuspid Valve Insufficiency/epidemiology , Tricuspid Valve Insufficiency/physiopathology , Cross-Sectional Studies , Cerebrovascular Disorders/epidemiology , Cerebrovascular Disorders/etiology , Child, Preschool , Young Adult , Infant , Kidney Diseases/etiology , Kidney Diseases/epidemiology , Kidney Diseases/physiopathology , Echocardiography , Adult , Follow-Up Studies , Prognosis
6.
Am J Physiol Renal Physiol ; 326(5): F768-F779, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38450435

ABSTRACT

Mitochondria are essential organelles in the human body, serving as the metabolic factory of the whole organism. When mitochondria are dysfunctional, it can affect all organs of the body. The kidney is rich in mitochondria, and its function is closely related to the development of kidney diseases. Studying the relationship between mitochondria and kidney disease progression is of great interest. In the past decade, scientists have made inspiring progress in investigating the role of mitochondria in the pathophysiology of renal diseases. This article discusses various mechanisms for maintaining mitochondrial quality, including mitochondrial energetics, mitochondrial biogenesis, mitochondrial dynamics, mitochondrial DNA repair, mitochondrial proteolysis and the unfolded protein response, mitochondrial autophagy, mitochondria-derived vesicles, and mitocytosis. The article also highlights the cross talk between mitochondria and other organelles, with a focus on kidney diseases. Finally, the article concludes with an overview of mitochondria-related clinical research.


Subject(s)
Kidney Diseases , Mitochondria , Humans , Mitochondria/metabolism , Mitochondria/pathology , Kidney Diseases/physiopathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Animals , Kidney/metabolism , Kidney/physiopathology , Kidney/pathology , Energy Metabolism , Autophagy , Mitochondrial Dynamics , Mitophagy , Unfolded Protein Response , Organelle Biogenesis
7.
J Med Ultrason (2001) ; 51(2): 159-168, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38206492

ABSTRACT

PURPOSE: Renal congestion is a therapeutic target in congestive heart failure. However, its detailed evaluation in a clinical setting is challenging. This study sought to assess renal congestion impairment using superb microvascular imaging (SMI), a simple and accessible method. METHODS: Dahl salt-sensitive rats, used as a model for congestive heart failure, underwent central venous pressure (CVP) measurements. Renal congestion was evaluated through measurements of renal medullary pressure (RMP) and assessment of renal perfusion using contrast-enhanced ultrasonography at both the early (control group) and heart failure phases (HF group). All rats were assessed with SMI. The region of interest (ROI) was set in interlobular vessels, interlobar vessels, and a combination of these areas. The area ratio was calculated from the color pixel count in the ROI divided by the total pixel count in the ROI. Intrarenal perfusion index (IRPI) was defined as (maximum area ratio-minimum area ratio) / maximum area ratio. RESULTS: There were no significant differences in renal function and left ventricular ejection fraction between the two groups. CVP, time-to-peak (TTP) in the medulla, and RMP were higher in the HF group than in the control group. In the HF group, IRPI, evaluated in the interlobular vessels, was significantly higher than in the control group. IRPI was positively correlated with TTP in the medulla (p = 0.028, R = 0.60) and RMP (p < 0.001, R = 0.84), indicating that IRPI reflected renal congestion. CONCLUSIONS: IRPI is a useful tool for assessing renal congestion in rats with congestive heart failure.


Subject(s)
Disease Models, Animal , Heart Failure , Kidney , Rats, Inbred Dahl , Animals , Heart Failure/diagnostic imaging , Heart Failure/physiopathology , Rats , Male , Kidney/blood supply , Kidney/diagnostic imaging , Ultrasonography/methods , Microvessels/diagnostic imaging , Microvessels/physiopathology , Contrast Media , Renal Circulation , Kidney Diseases/diagnostic imaging , Kidney Diseases/physiopathology
8.
Stem Cell Res Ther ; 14(1): 336, 2023 11 19.
Article in English | MEDLINE | ID: mdl-37981699

ABSTRACT

BACKGROUND: Kidney organoids derived from human pluripotent stem cells (HiPSCs) hold huge applications for drug screening, disease modeling, and cell transplanting therapy. However, these applications are limited since kidney organoid cannot maintain complete morphology and function like human kidney. Kidney organoids are not well differentiated since the core of the organoid lacked oxygen, nutrition, and vasculature, which creates essential niches. Hypoxia-inducible factor-1 α (HIF-1α) serves as a critical regulator in vascularization and cell survival under hypoxia environment. Less is known about the role of HIF-1α in kidney organoids in this regard. This study tried to investigate the effect of HIF-1α in kidney organoid vascularization and related disease modeling. METHODS: For the vascularization study, kidney organoids were generated from human induced pluripotent stem cells. We overexpressed HIF-1α via plasmid transfection or treated DMOG (Dimethyloxallyl Glycine, an agent for HIF-1α stabilization and accumulation) in kidney progenitor cells to detect the endothelium. For the disease modeling study, we treated kidney organoid with cisplatin under hypoxia environment, with additional HIF-1α transfection. RESULT: HIF-1α overexpression elicited kidney organoid vascularization. The endothelial cells and angiotool analysis parameters were increased in HIF-1α plasmid-transfected and DMOG-treated organoids. These angiogenesis processes were partially blocked by VEGFR inhibitors, semaxanib or axitinib. Cisplatin-induced kidney injury (Cleaved caspase 3) was protected by HIF-1α through the upregulation of CD31 and SOD2. CONCLUSION: We demonstrated that HIF-1α elicited the process of kidney organoid vascularization and protected against cisplatin-induced kidney organoid injury in hypoxia environment.


Subject(s)
Angiogenesis , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney , Models, Biological , Organoids , Organoids/blood supply , Organoids/metabolism , Kidney/metabolism , Multipotent Stem Cells , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Humans , Plasmids/genetics , Gene Expression , Stem Cells/cytology , Stem Cells/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Angiogenesis Inhibitors/pharmacology , Angiogenesis/drug effects , Angiogenesis/physiopathology , Axitinib/pharmacology , Cells, Cultured , Cisplatin/pharmacology , Cell Hypoxia , Kidney Diseases/physiopathology
9.
EBioMedicine ; 90: 104537, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37001235

ABSTRACT

BACKGROUND: Observational studies have investigated the effect of serum lipids on kidney function, but these findings are limited by confounding, reverse causation and have reported conflicting results. Mendelian randomization (MR) studies address this confounding problem. However, they have been conducted mostly in European ancestry individuals. We, therefore, set out to investigate the effect of lipid traits on the estimated glomerular filtration rate (eGFR) based on serum creatinine in individuals of African ancestry. METHODS: We used the two-sample and multivariable Mendelian randomization (MVMR) approaches; in which instrument variables (IV's) for the predictor (lipid traits) were derived from summary-level data of a meta-analyzed African lipid GWAS (MALG, n = 24,215) from the African Partnership for Chronic Disease Research (APCDR) (n = 13,612) & the Africa Wits-IN-DEPTH partnership for Genomics studies (AWI-Gen) dataset (n = 10,603). The outcome IV's were computed from the eGFR summary-level data of African-ancestry individuals within the Million Veteran Program (n = 57,336). A random-effects inverse variance method was used in our primary analysis, and pleiotropy was adjusted for using robust and penalized sensitivity testing. The lipid predictors for the MVMR were high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, and triglycerides (TG). FINDINGS: We found a significant causal association between genetically predicted low-density lipoprotein (LDL) cholesterol and eGFR in African ancestry individuals ß = 1.1 (95% CI [0.411-1.788]; p = 0.002). Similarly, total cholesterol (TC) showed a significant causal effect on eGFR ß = 1.619 (95% CI [0.412-2.826]; p = 0.009). However, the IVW estimate showed that genetically predicted HDL-C ß = -0.164, (95% CI = [-1.329 to 1.00]; p = 0.782), and TG ß = -0.934 (CI = [-2.815 to 0.947]; p = 0.33) were not significantly causally associated with the risk of eGFR. In the multivariable analysis inverse-variance weighted (MVIVW) method, there was evidence for a causal association between LDL and eGFR ß = 1.228 (CI = [0.477-1.979]; p = 0.001). A significant causal effect of Triglycerides (TG) on eGFR in the MVIVW analysis ß = -1.3 ([-2.533 to -0.067]; p = 0.039) was observed as well. All the causal estimates reported reflect a unit change in the outcome per a 1 SD increase in the exposure. HDL showed no evidence of a significant causal association with eGFR in the MVIVW method (ß = -0.117 (95% CI [-1.252 to 0.018]; p = 0.840)). We found no evidence of a reverse causal impact of eGFR on serum lipids. All our sensitivity analyses indicated no strong evidence of pleiotropy or heterogeneity between our instrumental variables for both the forward and reverse MR analysis. INTERPRETATION: In this African ancestry population, genetically predicted higher LDL-C and TC are causally associated with higher eGFR levels, which may suggest that the relationship between LDL, TC and kidney function may be U-shaped. And as such, lowering LDL_C does not necessarily improve risk of kidney disease. This may also imply the reason why LDL_C is seen to be a poorer predictor of kidney function compared to HDL. In addition, this further supports that more work is warranted to confirm the potential association between lipid traits and risk of kidney disease in individuals of African Ancestry. FUNDING: Wellcome (220740/Z/20/Z).


Subject(s)
African People , Kidney Diseases , Kidney , Lipids , Humans , African People/genetics , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Genome-Wide Association Study , Glomerular Filtration Rate/physiology , Kidney/physiopathology , Kidney Diseases/blood , Kidney Diseases/ethnology , Kidney Diseases/genetics , Kidney Diseases/physiopathology , Lipids/blood , Lipids/genetics , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Random Allocation , Risk Factors , Triglycerides/blood
12.
Comput Math Methods Med ; 2022: 2484626, 2022.
Article in English | MEDLINE | ID: mdl-35295198

ABSTRACT

Background: Glucocorticoids are the most effective anti-inflammatory and immunosuppressive drugs used to treat patients with renal disease. This study pooled the current evidence of the efficacy of Glucocorticoids and Glucocorticoid-induced hyperglycaemia in renal disease. Methods: We conducted a systematic literature search on PubMed, Cochrane Central, and Web of Science for relevant randomized controlled trials (RCTs) up to September 1, 2021. The meta-analysis, sensitivity analysis and bias analysis were performed using Review Manager 5. 3. Results: In this study, seven RCTs with 797 patients were included in our analysis. The analysis revealed that glucocorticoids had a certain alleviating effect on the reduction of renal function. (risk ratio [RR] 0.49 95% confidence interval [Cl] 0. 28 to 0.85, p =0.01) and reduction of proteinuria (weight mean difference [WMD] -0.43; 95% CI -0.57 to-0.28) when compared with the control group. Patients receiving glucocorticoids therapy did not have an increased risk of developing new-onset diabetes mellitus or impaired glucose tolerance. (RR 3.76 95% CI 0.54 to 26.10, p =0.18). For other safety outcomes, glucocorticoids therapy did not increase risk of respiratory infections (RR 1.63, 95% CI 0. 69to3. 89, p =0.27) and Gastrointestinal SAEs is relatively controversial (RR 1.10, 95% CI 0.32 to 3.79, p =0.88). Discussion. In conclusion, current clinical evidence indicates that glucocorticoids is efficacious and safe to renal disease compared with control. Further research comparing long-term glucocorticoids use is needed.


Subject(s)
Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use , Hyperglycemia/chemically induced , Kidney Diseases/drug therapy , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/therapeutic use , Blood Glucose/drug effects , Blood Glucose/metabolism , Computational Biology , Humans , Hyperglycemia/blood , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Kidney Diseases/physiopathology , Kidney Function Tests , Proteinuria/drug therapy , Randomized Controlled Trials as Topic , Risk Factors , Safety
13.
Toxins (Basel) ; 14(2)2022 02 17.
Article in English | MEDLINE | ID: mdl-35202173

ABSTRACT

Recent evidence has highlighted the importance of the gut microbiota in the pathophysiology of kidney diseases [...].


Subject(s)
Bacterial Toxins/toxicity , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/physiopathology , Gastrointestinal Microbiome/drug effects , Kidney Diseases/chemically induced , Kidney Diseases/physiopathology , Uremic Toxins/toxicity , Humans
14.
Clin Sci (Lond) ; 136(3): 239-256, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35129198

ABSTRACT

Renovascular disease (RVD) remains a common etiology of secondary hypertension. Recent clinical trials revealed unsatisfactory therapeutic outcomes of renal revascularization, leading to extensive investigation to unravel key pathophysiological mechanisms underlying irreversible functional loss and structural damage in the chronically ischemic kidney. Research studies identified complex interactions among various players, including inflammation, fibrosis, mitochondrial injury, cellular senescence, and microvascular remodeling. This interplay resulted in a shift of our understanding of RVD from a mere hemodynamic disorder to a pro-inflammatory and pro-fibrotic pathology strongly influenced by systemic diseases like metabolic syndrome (MetS), hypertension, diabetes mellitus, and hyperlipidemia. Novel diagnostic approaches have been tested for early detection and follow-up of RVD progression, using new imaging techniques and biochemical markers of renal injury and dysfunction. Therapies targeting some of the pathological pathways governing the development of RVD have shown promising results in animal models, and a few have moved from bench to clinical research. This review summarizes evolving understanding in chronic ischemic kidney injury.


Subject(s)
Ischemia , Kidney Diseases/physiopathology , Kidney/blood supply , Animals , Fibrosis , Humans , Hypertension, Renovascular/etiology , Inflammation , Kidney/physiopathology , Kidney Diseases/etiology , Renal Artery Obstruction , Renal Circulation
15.
Sci Rep ; 12(1): 1815, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110648

ABSTRACT

Citrate has been proposed as anticoagulation of choice in continuous renal replacement therapy (CRRT). However, little is known about the pharmacokinetics (PK) and metabolism of citrate in liver failure patients who require CRRT with regional citrate anticoagulation (RCA). This prospective clinical PK study was conducted at King Chulalongkorn Memorial Hospital between July 2019 to April 2021, evaluating seven acute liver failure (ALF) and seven acute-on-chronic liver failure (ACLF) patients who received CRRT support utilizing RCA as an anticoagulant at a citrate dose of 3 mmol/L. For evaluation of the citrate PK, we delivered citrate for 120 min and then stopped for a further 120 min. Total body clearance of citrate was 152.5 ± 50.9 and 195.6 ± 174.3 mL/min in ALF and ACLF, respectively. The ionized calcium, ionized magnesium, and pH slightly decreased after starting citrate infusion and gradually increased to baseline after stopping citrate infusion. Two of the ACLF patients displayed citrate toxicity during citrate infusion, while, no ALF patient had citrate toxicity. In summary, citrate clearance was significantly decreased in critically ill ALF and ACLF patients receiving CRRT. Citrate use as an anticoagulation in these patients is of concern for the risk of citrate toxicity.


Subject(s)
Anticoagulants/pharmacokinetics , Citric Acid/pharmacokinetics , Continuous Renal Replacement Therapy , Kidney Diseases/therapy , Liver Failure, Acute/therapy , Liver/metabolism , Acute-On-Chronic Liver Failure/blood , Acute-On-Chronic Liver Failure/diagnosis , Acute-On-Chronic Liver Failure/physiopathology , Acute-On-Chronic Liver Failure/therapy , Adult , Aged , Aged, 80 and over , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Citric Acid/administration & dosage , Citric Acid/adverse effects , Continuous Renal Replacement Therapy/adverse effects , Critical Illness , Female , Humans , Kidney Diseases/blood , Kidney Diseases/diagnosis , Kidney Diseases/physiopathology , Liver/physiopathology , Liver Failure, Acute/blood , Liver Failure, Acute/diagnosis , Liver Failure, Acute/physiopathology , Male , Metabolic Clearance Rate , Middle Aged , Prospective Studies , Treatment Outcome
16.
Life Sci ; 295: 120423, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35196530

ABSTRACT

Hepatocyte growth factor (HGF) has been proved to protect the liver against α-naphthylisothiocyanate (ANIT)-induced cholestasis by acting as an antioxidant agent and redirecting toxic biliary solutes towards blood for urinary excretion. However, this may represent an additional potential risk for kidney integrity, which is already compromised by the cholestatic process itself (cholemic nephropathy). Therefore, in the present work, we studied the renal damage caused by ANIT-induced cholestasis and whether it is aggravated or, on the contrary, counteracted by HGF; if the latter holds, the involvement of its antioxidant properties will be ascertained. ANIT-induced cholestatic deleterious renal effects were corroborated by the presence of urine bile salts, impairment of renal function, and the alterations of renal damage markers, such as HSP72, creatinine clearance, and albuminuria. HGF fully reverted all these, and the cast formation in the tubules was significantly decreased. These findings were associated with the control of renal oxidative stress. In summary, despite HGF enhancing the overload of potentially harmful biliary constituents that the kidney should remove from the bloodstream as an alternative depuration organ in cholestasis, it simultaneously protects the kidney from this damage by counteracting the prooxidant effects resulting from this harmful exposure.


Subject(s)
Cholestasis/drug therapy , Hepatocyte Growth Factor/pharmacology , Kidney Diseases/physiopathology , 1-Naphthylisothiocyanate/adverse effects , 1-Naphthylisothiocyanate/pharmacology , Animals , Antioxidants/pharmacology , Bile Acids and Salts/metabolism , Bile Ducts/physiopathology , Cholestasis/blood , Cholestasis/metabolism , Disease Models, Animal , Hepatocyte Growth Factor/metabolism , Kidney/metabolism , Kidney Diseases/metabolism , Liver/metabolism , Male , Mice , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
17.
Toxicol Lett ; 359: 10-21, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35114312

ABSTRACT

Organic anion transporters 1 (OAT1) and OAT3 are responsible for transporting adefovir (ADV) into renal tubular epithelial cells. Our previous research found that ADV accumulated in the renal interstitium and caused renal interstitial fibrosis when Oat1/3 were inhibited by OATs inhibitor probenecid for long-term. Mast cells (MCs) in the interstitial space are considered to be key drivers of renal fibrosis. The current work investigated the effect of ADV on MCs in vitro and during the development of interstitial fibrosis in rats. Results indicate that ADV triggers chymase release from cultured RBL-2H3 mast cells in a time-and concentration-dependent manner. Angiotensin II (Ang II) in renal interstitium is generated mainly by chymase, renin and other products released from MCs, and has a direct effect on fibrosis through the angiotensin receptor. The concentrations of Ang II and fibrosis was significantly increased after administration of ADV alone or with probenecid for 4 weeks. The MCs membrane stabilizer sodium cromoglycate (SCG) and the angiotensin receptor antagonist Valsartan (VAL) could ameliorate ADV-induced nephrotoxicity. Additionally, SCG or VAL could reduce the accumulation of ADV in the renal interstitium by upregulating the expression of Oat1/3 and multidrug resistance-associated protein 4. Therefore, ADV accumulation in the renal interstitium could promote the degranulation of interstitial MCs and drive the development of renal fibrosis. SCG or VAL could ameliorate ADV-associated fibrosis by decreasing degranulation of MCs and accelerating renal clearance of ADV.


Subject(s)
Adenine/analogs & derivatives , Adenine/toxicity , Cell Degranulation/drug effects , Fibrosis/chemically induced , Kidney Diseases/chemically induced , Mast Cells/drug effects , Organophosphonates/toxicity , Adenine/blood , Animals , Disease Models, Animal , Fibrosis/physiopathology , Humans , Kidney Diseases/physiopathology , Kidney Tubules/drug effects , Male , Organophosphonates/blood , Rats
18.
Nutrients ; 14(2)2022 Jan 07.
Article in English | MEDLINE | ID: mdl-35057425

ABSTRACT

The aim of this study was to analyse the association of diet with arterial stiffness and vascular ageing in a Caucasian population with intermediate cardiovascular risk. We recruited 2475 individuals aged 35-75 years with intermediate cardiovascular risk. Brachial-ankle pulse wave velocity (baPWV) was measured using a VaSera VS-1500® device. Vascular ageing was defined in two steps. Step 1: The 20 individuals who presented kidney disease, peripheral arterial disease, or heart failure were classified as early vascular ageing (EVA). Step 2: The individuals with percentiles by age and sex above the 90th percentile of baPWV among the participants of this study were classified as EVA, and the rest of the individuals were classified as non-EVA. The diet of the participants was analysed with two questionnaires: (1) the diet quality index (DQI) questionnaire and (2) the Mediterranean diet (MD) adherence questionnaire. The mean age of the sample was 61.34 ± 7.70 years, and 61.60% were men. Adherence to the MD was 53.30%. The DQI was 54.90%. Of the entire sample, 10.70% (11.15% of the men and 9.95% of the women) were EVA. In the multiple linear regression analysis, for each additional point in the DQI questionnaire, there was a decrease of -0.081 (95%CI (confidence intervals) -0.105--0.028) in baPWV; in the MD adherence questionnaire, there was a decrease of -0.052 (95%CI -0141--0.008). When performing the analysis, separated by sex, the association remained significant in men but not in women. In the logistic regression analysis, there was an increase in MD adherence and a decrease in the probability of presenting EVA, both with the DQI questionnaire (OR (odds ratio) = 0.65; 95%CI 0.50-0.84) and with the MD adherence questionnaire (OR = 0.75; 95%CI 0.58-0.97). In the analysis by sex, the association was only maintained in men (with DQI, OR = 0.54; 95%CI 0.37-0.56) (with MD, OR = 0.72; 95%CI 0.52-0.99). The results of this study suggest that a greater score in the DQI and MD adherence questionnaires is associated with lower arterial stiffness and a lower probability of presenting EVA. In the analysis by sex, this association is only observed in men.


Subject(s)
Aging/physiology , Arteries/physiology , Eating , Heart Disease Risk Factors , Vascular Stiffness/physiology , Adult , Age Factors , Aged , Ankle Brachial Index/instrumentation , Confidence Intervals , Cross-Sectional Studies , Diet Records , Diet, Mediterranean/statistics & numerical data , Female , Heart Failure/physiopathology , Humans , Kidney Diseases/physiopathology , Male , Middle Aged , Odds Ratio , Peripheral Arterial Disease/physiopathology , Pulse Wave Analysis/instrumentation , Regression Analysis , Sex Factors , White People
19.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-35064074

ABSTRACT

The human Sec61 complex is a widely distributed and abundant molecular machine. It resides in the membrane of the endoplasmic reticulum to channel two types of cargo: protein substrates and calcium ions. The SEC61A1 gene encodes for the pore-forming Sec61α subunit of the Sec61 complex. Despite their ubiquitous expression, the idiopathic SEC61A1 missense mutations p.V67G and p.T185A trigger a localized disease pattern diagnosed as autosomal dominant tubulointerstitial kidney disease (ADTKD-SEC61A1). Using cellular disease models for ADTKD-SEC61A1, we identified an impaired protein transport of the renal secretory protein renin and a reduced abundance of regulatory calcium transporters, including SERCA2. Treatment with the molecular chaperone phenylbutyrate reversed the defective protein transport of renin and the imbalanced calcium homeostasis. Signal peptide substitution experiments pointed at targeting sequences as the cause for the substrate-specific impairment of protein transport in the presence of the V67G or T185A mutations. Similarly, dominant mutations in the signal peptide of renin also cause ADTKD and point to impaired transport of this renal hormone as important pathogenic feature for ADTKD-SEC61A1 patients as well.


Subject(s)
Phenylbutyrates/pharmacology , Renin/metabolism , SEC Translocation Channels/genetics , Calcium/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Kidney Diseases/physiopathology , Molecular Chaperones/metabolism , Mutation, Missense , Phenylbutyrates/metabolism , Polycystic Kidney Diseases , Protein Transport/genetics , Renin/genetics , SEC Translocation Channels/chemistry , SEC Translocation Channels/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
20.
J Extracell Vesicles ; 11(1): e12181, 2022 01.
Article in English | MEDLINE | ID: mdl-35064766

ABSTRACT

Urinary extracellular vesicles (uEVs) are emerging as non-invasive biomarkers for various kidney diseases, but it is unknown how differences in nephron mass impact uEV excretion. To address this, uEV excretion was measured before and after human kidney donor nephrectomy and rat nephrectomy. In male and female donors, uEVs were quantified in cell-free spot and 24-h urine samples using nanoparticle tracking analysis (NTA), EVQuant, and CD9-time-resolved fluorescence immunoassay. Female donors had significantly lower total kidney volume (TKV) and excreted 49% fewer uEVs than male donors. uEV excretion correlated positively with estimated glomerular filtration rate (eGFR), creatinine clearance, and TKV (R's between 0.6 and 0.7). uEV excretion rate could also be predicted from spot urines after multiplying spot uEV/creatinine by 24-h urine creatinine. Donor nephrectomy reduced eGFR by 36% ± 10%, but the excretion of uEVs by only 16% (CD9+ uEVs -37%, CD9- uEVs no decrease). Donor nephrectomy increased the podocyte marker WT-1 and the proximal tubule markers NHE3, NaPi-IIa, and cubilin in uEVs two- to four-fold when correcting for the nephrectomy. In rats, the changes in GFR and kidney weight correlated with the changes in uEV excretion rate (R = 0.46 and 0.60, P < 0.01). Furthermore, the estimated degree of hypertrophy matched the change in uEV excretion rate (1.4- to 1.5-fold after uninephrectomy and four-fold after 5/6th nephrectomy). Taken together, our data show that uEV excretion depends on nephron mass, and that nephrectomy reduces uEV excretion less than expected based on nephron loss due to compensatory hypertrophy. The major implication of our findings is that a measure for nephron mass or uEV excretion rate should be included when comparing uEV biomarkers between individuals.


Subject(s)
Extracellular Vesicles/metabolism , Nephrons/physiology , Animals , Biomarkers/urine , Female , Humans , Kidney/metabolism , Kidney/physiology , Kidney/surgery , Kidney Diseases/physiopathology , Kidney Diseases/urine , Male , Nephrectomy , Rats , Sex Factors , Tissue Donors , Urinalysis/standards , Urinary Bladder/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...