Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Gut Microbes ; 16(1): 2341449, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38686499

RESUMO

In today's industrialized society food consumption has changed immensely toward heightened red meat intake and use of artificial sweeteners instead of grains and vegetables or sugar, respectively. These dietary changes affect public health in general through an increased incidence of metabolic diseases like diabetes and obesity, with a further elevated risk for cardiorenal complications. Research shows that high red meat intake and artificial sweeteners ingestion can alter the microbial composition and further intestinal wall barrier permeability allowing increased transmission of uremic toxins like p-cresyl sulfate, indoxyl sulfate, trimethylamine n-oxide and phenylacetylglutamine into the blood stream causing an array of pathophysiological effects especially as a strain on the kidneys, since they are responsible for clearing out the toxins. In this review, we address how the burden of the Western diet affects the gut microbiome in altering the microbial composition and increasing the gut permeability for uremic toxins and the detrimental effects thereof on early vascular aging, the kidney per se and the blood-brain barrier, in addition to the potential implications for dietary changes/interventions to preserve the health issues related to chronic diseases in future.


Assuntos
Barreira Hematoencefálica , Microbioma Gastrointestinal , Rim , Insuficiência Renal Crônica , Humanos , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/microbiologia , Insuficiência Renal Crônica/fisiopatologia , Barreira Hematoencefálica/metabolismo , Rim/fisiopatologia , Rim/metabolismo , Animais , Toxinas Urêmicas/metabolismo , Dieta Ocidental/efeitos adversos
2.
J Intern Med ; 295(1): 79-90, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37827529

RESUMO

BACKGROUND: Chronic kidney disease (CKD) is an age-related disease that displays multiple features of accelerated ageing. It is currently unclear whether the two treatment options for end-stage kidney disease (dialysis and kidney transplantation [KT]) ameliorate the accelerated uremic ageing process. METHODS: Data on clinical variables and blood DNA methylation (DNAm) from CKD stage G3-G5 patients were used to estimate biological age based on blood biomarkers (phenotypic age [PA], n = 333), skin autofluorescence (SAF age, n = 199) and DNAm (Horvath, Hannum and PhenoAge clocks, n = 47). In the DNAm cohort, we also measured the change in biological age 1 year after the KT or initiation of dialysis. Healthy subjects recruited from the general population were included as controls. RESULTS: All three DNAm clocks indicated an increased biological age in CKD G5. However, PA and SAF age tended to produce implausibly large estimates of biological age in CKD G5. By contrast, DNAm age was 4.9 years (p = 0.005) higher in the transplantation group and 5.9 years (p = 0.001) higher in the dialysis group compared to controls. This age acceleration was significantly reduced 1 year after KT, but not after 1 year of dialysis. CONCLUSIONS: Kidney failure patients displayed an increased biological age as estimated by DNAm clocks compared to population-based controls. Our results suggest that KT, but not dialysis, partially reduces the age acceleration.


Assuntos
Transplante de Rim , Insuficiência Renal Crônica , Humanos , Lactente , Pré-Escolar , Diálise Renal , Envelhecimento/genética , Metilação de DNA , Insuficiência Renal Crônica/terapia , Epigênese Genética
3.
Kidney Int ; 103(6): 1120-1130, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36990215

RESUMO

Morphological alterations at the kidney filtration barrier increase intrinsic capillary wall permeability resulting in albuminuria. However, automated, quantitative assessment of these morphological changes has not been possible with electron or light microscopy. Here we present a deep learning-based approach for segmentation and quantitative analysis of foot processes in images acquired with confocal and super-resolution fluorescence microscopy. Our method, Automatic Morphological Analysis of Podocytes (AMAP), accurately segments podocyte foot processes and quantifies their morphology. AMAP applied to a set of kidney diseases in patient biopsies and a mouse model of focal segmental glomerulosclerosis allowed for accurate and comprehensive quantification of various morphometric features. With the use of AMAP, detailed morphology of podocyte foot process effacement was found to differ between categories of kidney pathologies, showed detailed variability between diverse patients with the same clinical diagnosis, and correlated with levels of proteinuria. AMAP could potentially complement other readouts such as various omics, standard histologic/electron microscopy and blood/urine assays for future personalized diagnosis and treatment of kidney disease. Thus, our novel finding could have implications to afford an understanding of early phases of kidney disease progression and may provide supplemental information in precision diagnostics.


Assuntos
Aprendizado Profundo , Glomerulosclerose Segmentar e Focal , Nefropatias , Podócitos , Camundongos , Animais , Podócitos/patologia , Glomérulos Renais/patologia , Rim/diagnóstico por imagem , Rim/patologia , Glomerulosclerose Segmentar e Focal/diagnóstico por imagem , Glomerulosclerose Segmentar e Focal/patologia , Nefropatias/diagnóstico por imagem , Nefropatias/patologia
4.
Nephrol Dial Transplant ; 38(2): 311-321, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-35767852

RESUMO

BACKGROUND: Immunoglobulin A nephropathy (IgAN) and its systemic variant IgA vasculitis (IgAV) damage the glomeruli, resulting in proteinuria, hematuria and kidney impairment. Dendrin is a podocyte-specific protein suggested to be involved in the pathogenesis of IgAN. Upon cell injury, dendrin translocates from the slit diaphragm to the nucleus, where it is suggested to induce apoptosis and cytoskeletal changes, resulting in proteinuria and accelerated disease progression in mice. Here we investigated gene and protein expression of dendrin in relation to clinical and histopathological findings to further elucidate its role in IgAN/IgAV. METHODS: Glomerular gene expression was measured using microarray on 30 IgAN/IgAV patients, 5 patients with membranous nephropathy (MN) and 20 deceased kidney donors. Dendrin was spatially evaluated on kidney tissue sections by immunofluorescence (IF) staining (IgAN patients, n = 4; nephrectomized kidneys, n = 3) and semi-quantified by immunogold electron microscopy (IgAN/IgAV patients, n = 21; MN, n = 5; living kidney donors, n = 6). Histopathological grading was performed according to the Oxford and Banff classifications. Clinical data were collected at the time of biopsy and follow-up. RESULTS: Dendrin mRNA levels were higher (P = .01) in IgAN patients compared with MN patients and controls and most prominently in patients with preserved kidney function and fewer chronic histopathological changes. Whereas IF staining did not differ between groups, immunoelectron microscopy revealed that a higher relative nuclear dendrin concentration in IgAN patients was associated with a slower annual progression rate and milder histopathological changes. CONCLUSION: Dendrin messenger RNA levels and relative nuclear protein concentrations are increased and associated with a more benign phenotype and progression in IgAN/IgAV patients.


Assuntos
Glomerulonefrite por IGA , Glomerulonefrite Membranosa , Vasculite por IgA , Camundongos , Animais , Glomerulonefrite por IGA/complicações , Glomérulos Renais/patologia , Proteínas do Tecido Nervoso/metabolismo , Glomerulonefrite Membranosa/metabolismo , Vasculite por IgA/complicações , Proteinúria/etiologia
5.
Clin Transl Med ; 12(2): e682, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35184400

RESUMO

RATIONALE: Vascular calcification is a prominent feature of late-stage diabetes, renal and cardiovascular disease (CVD), and has been linked to adverse events. Recent studies in patients reported that plasma levels of osteomodulin (OMD), a proteoglycan involved in bone mineralisation, associate with diabetes and CVD. We hypothesised that OMD could be implicated in these diseases via vascular calcification as a common underlying factor and aimed to investigate its role in this context. METHODS AND RESULTS: In patients with chronic kidney disease, plasma OMD levels correlated with markers of inflammation and bone turnover, with the protein present in calcified arterial media. Plasma OMD also associated with cardiac calcification and the protein was detected in calcified valve leaflets by immunohistochemistry. In patients with carotid atherosclerosis, circulating OMD was increased in association with plaque calcification as assessed by computed tomography. Transcriptomic and proteomic data showed that OMD was upregulated in atherosclerotic compared to control arteries, particularly in calcified plaques, where OMD expression correlated positively with markers of smooth muscle cells (SMCs), osteoblasts and glycoproteins. Immunostaining confirmed that OMD was abundantly present in calcified plaques, localised to extracellular matrix and regions rich in α-SMA+ cells. In vivo, OMD was enriched in SMCs around calcified nodules in aortic media of nephrectomised rats and in plaques from ApoE-/- mice on warfarin. In vitro experiments revealed that OMD mRNA was upregulated in SMCs stimulated with IFNγ, BMP2, TGFß1, phosphate and ß-glycerophosphate, and by administration of recombinant human OMD protein (rhOMD). Mechanistically, addition of rhOMD repressed the calcification process of SMCs treated with phosphate by maintaining their contractile phenotype along with enriched matrix organisation, thereby attenuating SMC osteoblastic transformation. Mechanistically, the role of OMD is exerted likely through its link with SMAD3 and TGFB1 signalling, and interplay with BMP2 in vascular tissues. CONCLUSION: We report a consistent association of both circulating and tissue OMD levels with cardiovascular calcification, highlighting the potential of OMD as a clinical biomarker. OMD was localised in medial and intimal α-SMA+ regions of calcified cardiovascular tissues, induced by pro-inflammatory and pro-osteogenic stimuli, while the presence of OMD in extracellular environment attenuated SMC calcification.


Assuntos
Proteínas da Matriz Extracelular/farmacologia , Músculo Liso/efeitos dos fármacos , Osteogênese/genética , Proteoglicanas/farmacologia , Calcificação Vascular/etiologia , Análise de Variância , Estudos de Coortes , Estudos Transversais , Proteínas da Matriz Extracelular/metabolismo , Humanos , Modelos Lineares , Músculo Liso/fisiologia , Países Baixos , Osteogênese/fisiologia , Estudos Prospectivos , Proteoglicanas/metabolismo , Estatísticas não Paramétricas , Suécia , Calcificação Vascular/genética
6.
Sci Rep ; 12(1): 470, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013499

RESUMO

Chronic kidney disease (CKD) is an emerging public health priority associated with high mortality rates and demanding treatment regimens, including life-style changes, medications or even dialysis or renal transplantation. Unavoidably, the uremic milieu disturbs homeostatic processes such as DNA methylation and other vital gene regulatory mechanisms. Here, we aimed to investigate how dialysis or kidney transplantation modifies the epigenome-wide methylation signature over 12 months of treatment. We used the Infinium HumanMethylation450 BeadChip on whole blood samples from CKD-patients undergoing either dialysis (n = 11) or kidney transplantation (n = 12) and 24 age- and sex-matched population-based controls. At baseline, comparison between patients and controls identified several significant (PFDR < 0.01) CpG methylation differences in genes with functions relevant to inflammation, cellular ageing and vascular calcification. Following 12 months, the global DNA methylation pattern of patients approached that seen in the control group. Notably, 413 CpG sites remained differentially methylated at follow-up in both treatment groups compared to controls. Together, these data indicate that the uremic milieu drives genome-wide methylation changes that are partially reversed with kidney failure replacement therapy. Differentially methylated CpG sites unaffected by treatment may be of particular interest as they could highlight candidate genes for kidney disease per se.


Assuntos
Epigênese Genética , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/terapia , Adulto , Idoso , Estudos de Casos e Controles , Ilhas de CpG , Metilação de DNA , Epigenoma , Feminino , Estudo de Associação Genômica Ampla , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Diálise Renal , Terapia de Substituição Renal
7.
Front Endocrinol (Lausanne) ; 13: 1046392, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699036

RESUMO

Background: Disturbances in magnesium homeostasis are common in patients with chronic kidney disease (CKD) and are associated with increased mortality. The kidney is a key organ in maintaining normal serum magnesium concentrations. To this end, fractional excretion of magnesium (FEMg) increases as renal function declines. Despite recent progress, the hormonal regulation of renal magnesium handling is incompletely understood. Fibroblast Growth Factor 23 (FGF23) is a phosphaturic hormone that has been linked to renal magnesium handling. However, it has not yet been reported whether FGF23 is associated with renal magnesium handling in CKD patients. Methods: The associations between plasma FGF23 levels, plasma and urine magnesium concentrations and FEMg was investigated in a cross-sectional cohort of 198 non-dialysis CKD patients undergoing renal biopsy. Results: FGF23 was significantly correlated with FEMg (Pearson's correlation coefficient = 0.37, p<0.001) and urinary magnesium (-0.14, p=0.04), but not with plasma magnesium. The association between FGF23 and FEMg remained significant after adjusting for potential confounders, including estimated glomerular filtration rate (eGFR), parathyroid hormone and 25-hydroxyvitamin D. Conclusions: We report that plasma FGF23 is independently associated with measures of renal magnesium handling in a cohort of non-dialysis CKD patients. A potential causal relationship should be investigated in future studies.


Assuntos
Magnésio , Insuficiência Renal Crônica , Humanos , Fator de Crescimento de Fibroblastos 23 , Estudos Transversais , Fatores de Crescimento de Fibroblastos/metabolismo , Rim/metabolismo , Insuficiência Renal Crônica/complicações
8.
Antioxid Redox Signal ; 35(17): 1426-1448, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34006115

RESUMO

Significance: Chronic kidney disease (CKD) can be regarded as a burden of lifestyle disease that shares common underpinning features and risk factors with the aging process; it is a complex constituted by several adverse components, including chronic inflammation, oxidative stress, early vascular aging, and cellular senescence. Recent Advances: A systemic approach to tackle CKD, based on mitigating the associated inflammatory, cell stress, and damage processes, has the potential to attenuate the effects of CKD, but it also preempts the development and progression of associated morbidities. In effect, this will enhance health span and compress the period of morbidity. Pharmacological, nutritional, and potentially lifestyle-based interventions are promising therapeutic avenues to achieve such a goal. Critical Issues: In the present review, currents concepts of inflammation and oxidative damage as key patho-mechanisms in CKD are addressed. In particular, potential beneficial but also adverse effects of different systemic interventions in patients with CKD are discussed. Future Directions: Senotherapeutics, the nuclear factor erythroid 2-related factor 2-kelch-like ECH-associated protein 1 (NRF2-KEAP1) signaling pathway, the endocrine klotho axis, inhibitors of the sodium-glucose cotransporter 2 (SGLT2), and live bio-therapeutics have the potential to reduce the burden of CKD and improve quality of life, as well as morbidity and mortality, in this fragile high-risk patient group. Antioxid. Redox Signal. 35, 1426-1448.


Assuntos
Qualidade de Vida , Insuficiência Renal Crônica , Humanos , Inflamação/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Diálise Renal , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/terapia
9.
Nat Commun ; 12(1): 2141, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33837218

RESUMO

Molecular characterization of the individual cell types in human kidney as well as model organisms are critical in defining organ function and understanding translational aspects of biomedical research. Previous studies have uncovered gene expression profiles of several kidney glomerular cell types, however, important cells, including mesangial (MCs) and glomerular parietal epithelial cells (PECs), are missing or incompletely described, and a systematic comparison between mouse and human kidney is lacking. To this end, we use Smart-seq2 to profile 4332 individual glomerulus-associated cells isolated from human living donor renal biopsies and mouse kidney. The analysis reveals genetic programs for all four glomerular cell types (podocytes, glomerular endothelial cells, MCs and PECs) as well as rare glomerulus-associated macula densa cells. Importantly, we detect heterogeneity in glomerulus-associated Pdgfrb-expressing cells, including bona fide intraglomerular MCs with the functionally active phagocytic molecular machinery, as well as a unique mural cell type located in the central stalk region of the glomerulus tuft. Furthermore, we observe remarkable species differences in the individual gene expression profiles of defined glomerular cell types that highlight translational challenges in the field and provide a guide to design translational studies.


Assuntos
Células Endoteliais/metabolismo , Mesângio Glomerular/metabolismo , Podócitos/metabolismo , Biossíntese de Proteínas/genética , Transcriptoma/fisiologia , Animais , Separação Celular , Biologia Computacional , Citometria de Fluxo , Heterogeneidade Genética , Mesângio Glomerular/citologia , Humanos , Masculino , Camundongos , RNA-Seq , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptores da Fosfolipase A2/genética , Análise de Célula Única , Especificidade da Espécie
10.
PLoS One ; 16(2): e0247623, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33626087

RESUMO

Patients with chronic kidney disease (CKD) suffer from vitamin K deficiency and are at high risk of vascular calcification (VC) and premature death. We investigated the association of functional vitamin K deficiency with all-cause mortality and whether this association is modified by the presence of VC in CKD stage 5 (CKD G5). Plasma dephosphorylated-uncarboxylated matrix Gla-protein (dp-ucMGP), a circulating marker of functional vitamin K deficiency, and other laboratory and clinical data were determined in 493 CKD G5 patients. VC was assessed in subgroups by Agatston scoring of coronary artery calcium (CAC) and aortic valve calcium (AVC). Backward stepwise regression did not identify dp-ucMGP as an independent determinant of VC. During a median follow-up of 42 months, 93 patients died. Each one standard deviation increment in dp-ucMGP was associated with increased risk of all-cause mortality (sub-hazard ratio (sHR) 1.17; 95% confidence interval, 1.01-1.37) adjusted for age, sex, cardiovascular disease, diabetes, body mass index, inflammation, and dialysis treatment. The association remained significant when further adjusted for CAC and AVC in sub-analyses (sHR 1.22, 1.01-1.48 and 1.27, 1.01-1.60, respectively). In conclusion, functional vitamin K deficiency associates with increased mortality risk that is independent of the presence of VC in patients with CKD G5.


Assuntos
Insuficiência Renal Crônica/complicações , Calcificação Vascular/complicações , Deficiência de Vitamina K/complicações , Vitamina K/sangue , Adulto , Idoso , Biomarcadores/sangue , Proteínas de Ligação ao Cálcio/sangue , Estudos de Coortes , Proteínas da Matriz Extracelular/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/mortalidade , Insuficiência Renal Crônica/patologia , Taxa de Sobrevida , Calcificação Vascular/sangue , Calcificação Vascular/mortalidade , Calcificação Vascular/patologia , Deficiência de Vitamina K/sangue , Deficiência de Vitamina K/mortalidade , Deficiência de Vitamina K/patologia , Proteína de Matriz Gla
11.
Kidney Int ; 99(4): 1010-1020, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33285146

RESUMO

In recent years, many light-microscopy protocols have been published for visualization of nanoscale structures in the kidney. These protocols present researchers with new tools to evaluate both foot process anatomy and effacement, as well as protein distributions in foot processes, the slit diaphragm and in the glomerular basement membrane. However, these protocols either involve the application of different complicated super resolution microscopes or lengthy sample preparation protocols. Here, we present a fast and simple, five-hour long procedure for three-dimensional visualization of kidney morphology on all length scales. The protocol combines optical clearing and tissue expansion concepts to produce a mild swelling, sufficient for resolving nanoscale structures using a conventional confocal microscope. We show that the protocol can be applied to visualize a wide variety of pathologic features in both mouse and human kidneys. Thus, our fast and simple protocol can be beneficial for conventional microscopic evaluation of kidney tissue integrity both in research and possibly in future clinical routines.


Assuntos
Glomérulos Renais , Rim , Animais , Rim/diagnóstico por imagem , Camundongos , Microscopia
12.
Nephrol Dial Transplant ; 35(12): 2059-2072, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-32853351

RESUMO

BACKGROUND: Diabetic nephropathy (DN) is the most common cause of end-stage renal disease, affecting ∼30% of the rapidly growing diabetic population, and strongly associated with cardiovascular risk. Despite this, the molecular mechanisms of disease remain unknown. METHODS: RNA sequencing (RNAseq) was performed on paired, micro-dissected glomerular and tubulointerstitial tissue from patients diagnosed with DN [n = 19, 15 males, median (range) age: 61 (30-85) years, chronic kidney disease stages 1-4] and living kidney donors [n = 20, 12 males, median (range) age: 56 (30-70) years]. RESULTS: Principal component analysis showed a clear separation between glomeruli and tubulointerstitium transcriptomes. Differential expression analysis identified 1550 and 4530 differentially expressed genes, respectively (adjusted P < 0.01). Gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses highlighted activation of inflammation and extracellular matrix (ECM) organization pathways in glomeruli, and immune and apoptosis pathways in tubulointerstitium of DN patients. Specific gene modules were associated with renal function in weighted gene co-expression network analysis. Increased messengerRNA (mRNA) expression of renal damage markers lipocalin 2 (LCN) and hepatitis A virus cellular receptor1 (HAVCR1) in the tubulointerstitial fraction was observed alongside higher urinary concentrations of the corresponding proteins neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) in DN patients. CONCLUSIONS: Here we present the first RNAseq experiment performed on paired glomerular and tubulointerstitial samples from DN patients. We show that prominent disease-specific changes occur in both compartments, including relevant cellular processes such as reorganization of ECM and inflammation (glomeruli) as well as apoptosis (tubulointerstitium). The results emphasize the potential of utilizing high-throughput transcriptomics to decipher disease pathways and treatment targets in this high-risk patient population.


Assuntos
Biomarcadores/análise , Diabetes Mellitus/fisiopatologia , Nefropatias Diabéticas/genética , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Transcriptoma , Adulto , Idoso , Idoso de 80 Anos ou mais , Biologia Computacional/métodos , Nefropatias Diabéticas/epidemiologia , Nefropatias Diabéticas/patologia , Feminino , Receptor Celular 1 do Vírus da Hepatite A/genética , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Humanos , Testes de Função Renal , Glomérulos Renais/patologia , Túbulos Renais/patologia , Lipocalina-2/genética , Lipocalina-2/metabolismo , Masculino , Pessoa de Meia-Idade , Suécia/epidemiologia
13.
Sci Rep ; 10(1): 6586, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313061

RESUMO

Matrix Gla protein (MGP) is a potent inhibitor of vascular calcification (VC) and requires carboxylation by vitamin K to exert calcification inhibition. Chronic kidney disease (CKD) patients undergo early vascular aging often involving extensive VC. The present cross-sectional study investigated the association between circulating dp-ucMGP levels, MGP expression in vascular tissue and MGP polymorphisms. In 141 CKD stage 5 patients, CAC score was significantly increased in the highest tertile of dp-ucMGP (p = 0.002), and a high medial VC score was associated with elevated dp-ucMGP levels. MGP vascular expression was associated with increased circulating dp-ucMGP and CAC scores. MGP SNP analysis revealed that patients homozygous for the C allele of the rs1800801 variant had a higher CAC score (median 15 [range 0-1312]) compared to patients carrying a T allele (median 0 [range 0-966] AU). These results indicate that plasma levels of dp-ucMGP are an independent predictor of increased VC in CKD5 patients and correlate with both higher CAC scores and degree of medial calcification. Additionally, high vascular expression of MGP was associated with higher CAC scores and plasma dp-ucMGP levels. Taken together, our results support that MGP is involved in the pathogenesis of VC.


Assuntos
Proteínas de Ligação ao Cálcio/sangue , Proteínas da Matriz Extracelular/sangue , Falência Renal Crônica/sangue , Insuficiência Renal Crônica/sangue , Calcificação Vascular/sangue , Adulto , Biomarcadores/sangue , Proteínas de Ligação ao Cálcio/genética , Espessura Intima-Media Carotídea , Proteínas da Matriz Extracelular/genética , Feminino , Regulação da Expressão Gênica/genética , Humanos , Falência Renal Crônica/genética , Falência Renal Crônica/patologia , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Fatores de Risco , Calcificação Vascular/genética , Calcificação Vascular/patologia , Proteína de Matriz Gla
14.
Toxins (Basel) ; 12(4)2020 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-32260373

RESUMO

Persistent low-grade inflammation and premature ageing are hallmarks of the uremic phenotype and contribute to impaired health status, reduced quality of life, and premature mortality in chronic kidney disease (CKD). Because there is a huge global burden of disease due to CKD, treatment strategies targeting inflammation and premature ageing in CKD are of particular interest. Several distinct features of the uremic phenotype may represent potential treatment options to attenuate the risk of progression and poor outcome in CKD. The nuclear factor erythroid 2-related factor 2 (NRF2)-kelch-like erythroid cell-derived protein with CNC homology [ECH]-associated protein 1 (KEAP1) signaling pathway, the endocrine phosphate-fibroblast growth factor-23-klotho axis, increased cellular senescence, and impaired mitochondrial biogenesis are currently the most promising candidates, and different pharmaceutical compounds are already under evaluation. If studies in humans show beneficial effects, carefully phenotyped patients with CKD can benefit from them.


Assuntos
Senilidade Prematura/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Rim/metabolismo , Insuficiência Renal Crônica/metabolismo , Toxinas Biológicas/metabolismo , Uremia/metabolismo , Senilidade Prematura/mortalidade , Senilidade Prematura/fisiopatologia , Animais , Biomarcadores/metabolismo , Nível de Saúde , Humanos , Inflamação/mortalidade , Inflamação/fisiopatologia , Inflamação/terapia , Rim/patologia , Rim/fisiopatologia , Fenótipo , Prognóstico , Qualidade de Vida , Insuficiência Renal Crônica/mortalidade , Insuficiência Renal Crônica/fisiopatologia , Insuficiência Renal Crônica/terapia , Fatores de Risco , Transdução de Sinais , Uremia/mortalidade , Uremia/fisiopatologia , Uremia/terapia
15.
Am J Physiol Renal Physiol ; 317(5): F1122-F1131, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31432706

RESUMO

Advanced chronic kidney disease (CKD) is characterized by a premature aging phenotype of multifactorial origin. Mitochondrial dysfunction is prevalent in CKD and has been proposed as a major contributor to poor muscle function. Although the mitochondria-derived peptides (MDPs) humanin and mitochondrial open reading frame of 12S rRNA-c (MOTS-c) are involved in cell survival, suppression of apoptosis, and glucose control, the implications of MDP in CKD are unknown. We investigated humanin and MOTS-c protein expression in skeletal muscle and serum levels in CKD at stage 5 (glomerular filtration rate: <15 ml/min) patients and age-matched controls with normal renal function. Whereas circulating levels of humanin were increased in CKD, local muscle expression was reduced. In contrast, MOTS-c levels were reduced in both skeletal muscle and serum in CKD. Humanin in serum correlated positively to circulating TNF levels. Reduced MDP levels in skeletal muscle were associated with lower mitochondrial density and evidence of oxidative stress. These results indicate a differential regulation of MDPs in CKD and suggest an alternative site for humanin production than skeletal muscle in the uremic milieu. MDP levels were linked to systemic inflammation and evidence of oxidative stress in the muscle, two hallmark features of premature aging and uremia.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias Musculares/metabolismo , Proteínas Mitocondriais/metabolismo , Músculo Esquelético/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Insuficiência Renal Crônica/metabolismo , Adulto , Idoso , Regulação para Baixo , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Mitocondriais/genética , Fator 2 Relacionado a NF-E2/genética , Adulto Jovem
16.
Comput Struct Biotechnol J ; 17: 721-729, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31303976

RESUMO

Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by progressive vascular disease, systemic inflammation, muscle wasting and frailty. The predominant early vascular ageing (EVA) process mediated by medial vascular calcification (VC) results in a marked discrepancy between chronological and biological vascular age in CKD. Though the exact underlying mechanisms of VC and EVA are not fully elucidated, accumulating evidence indicates that cellular senescence - and subsequent chronic inflammation through the senescence-associated secretary phenotype (SASP) - plays a fundamental role in its initiation and progression. In this review, we discuss the pathophysiological links between senescence and the EVA process in CKD, with focus on cellular senescence and media VC, and potential anti-ageing therapeutic strategies of senolytic drugs targeting cellular senescence and EVA in CKD.

17.
Diabetol Metab Syndr ; 11: 32, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31168327

RESUMO

BACKGROUND: Medial calcification in diabetes contributes to the arterial occlusive process occurring below the knee level. Adiponectin is an adipokine with atheroprotective properties and possible protective role against arterial calcification. The aim of the study was to investigate, in type 2 diabetes, the link between vascular expression and serum concentration of adiponectin and (1) peripheral arterial calcification and (2) lower limb occlusive arterial disease. METHODS: Scoring of peripheral vascular calcification and peripheral arterial occlusive disease, using CT-scan and color-duplex ultrasonography respectively, were conducted and explored in relation to serum adiponectin level in a cross sectional study of 197 patients with type 2 diabetes. Vascular adiponectin expression in the arterial wall of diabetic patients with and without medial calcification was evaluated by immunohistochemistry. RESULTS: Peripheral arterial calcification score was higher in patients with the highest adiponectin concentration. In a multivariate logistic regression analysis, an increase of 1 µg/mL of adiponectin was associated with a 22% increase of arterial calcification (adjusted OR = 1.22; 95% CI 1.03-1.44; p = 0.02). Arterial occlusive score was also higher in patients with adiponectin concentration > median (2.8 ± 4.8 vs 4.2 ± 5.7, p = 0.034). Immunohistochemical analyses showed a strong and specific staining of adiponectin in smooth muscle cells in calcified arteries, with a more pronounced expression of adiponectin in early stages of medial calcification. CONCLUSIONS: Peripheral arterial calcification is positively associated with circulating adiponectin levels in patients with type 2 diabetes, but vascular adiponectin expression is already observed at early stages of calcification. Adiponectin secretion could be a compensatory mechanism against the calcification process.Trial registration DIACART NCT number: NCT02431234. Registered 30 April 2015.

18.
Atherosclerosis ; 288: 175-185, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31109707

RESUMO

BACKGROUND AND AIMS: Unstable carotid atherosclerosis causes stroke, but methods to identify patients and lesions at risk are lacking. We recently found enrichment of genes associated with calcification in carotid plaques from asymptomatic patients. Here, we hypothesized that calcification represents a stabilising feature of plaques and investigated how macro-calcification, as estimated by computed tomography (CT), correlates with gene expression profiles in lesions. METHODS: Plaque calcification was measured in pre-operative CT angiographies. Plaques were sorted into high- and low-calcified, profiled with microarrays, followed by bioinformatic analyses. Immunohistochemistry and qPCR were performed to evaluate the findings in plaques and arteries with medial calcification from chronic kidney disease patients. RESULTS: Smooth muscle cell (SMC) markers were upregulated in high-calcified plaques and calcified plaques from symptomatic patients, whereas macrophage markers were downregulated. The most enriched processes in high-calcified plaques were related to SMCs and extracellular matrix (ECM) organization, while inflammation, lipid transport and chemokine signaling were repressed. These findings were confirmed in arteries with high medial calcification. Proteoglycan 4 (PRG4) was identified as the most upregulated gene in association with plaque calcification and found in the ECM, SMA+ and CD68+/TRAP + cells. CONCLUSIONS: Macro-calcification in carotid lesions correlated with a transcriptional profile typical for stable plaques, with altered SMC phenotype and ECM composition and repressed inflammation. PRG4, previously not described in atherosclerosis, was enriched in the calcified ECM and localized to activated macrophages and smooth muscle-like cells. This study strengthens the notion that assessment of calcification may aid evaluation of plaque phenotype and stroke risk.


Assuntos
Estenose das Carótidas/diagnóstico por imagem , Estenose das Carótidas/genética , Angiografia por Tomografia Computadorizada , Perfilação da Expressão Gênica , Músculo Liso Vascular/diagnóstico por imagem , Placa Aterosclerótica , Transcriptoma , Calcificação Vascular/diagnóstico por imagem , Calcificação Vascular/genética , Artérias Carótidas/diagnóstico por imagem , Artérias Carótidas/patologia , Estenose das Carótidas/complicações , Estenose das Carótidas/patologia , Humanos , Músculo Liso Vascular/patologia , Valor Preditivo dos Testes , Proteoglicanas/genética , Medição de Risco , Fatores de Risco , Ruptura Espontânea , Acidente Vascular Cerebral/etiologia , Suécia , Calcificação Vascular/complicações , Calcificação Vascular/patologia
19.
Cardiovasc Res ; 115(10): 1557-1566, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30597013

RESUMO

AIMS: Vascular calcification, a marker of increased cardiovascular risk, is an active process orchestrated by smooth muscle cells. Observational studies indicate that omega-3 fatty acids protect against vascular calcification, but the mechanisms are unknown. The G-protein coupled receptor ChemR23 transduces the resolution of inflammation induced by the omega-3-derived lipid mediator resolvin E1. ChemR23 also contributes to osteoblastic differentiation of stem cells and bone formation, but its role in vascular calcification is unknown. The aim of this study was to establish the role of ChemR23 in smooth muscle cell fate and calcification. METHODS AND RESULTS: Gene expression analysis in epigastric arteries derived from patients with chronic kidney disease and vascular calcification revealed that ChemR23 mRNA levels predicted a synthetic smooth muscle cell phenotype. Genetic deletion of ChemR23 in mice prevented smooth muscle cell de-differentiation. ChemR23-deficient smooth muscle cells maintained a non-synthetic phenotype and exhibited resistance to phosphate-induced calcification. Moreover, ChemR23-deficient mice were protected against vitamin D3-induced vascular calcification. Resolvin E1 inhibited smooth muscle cell calcification through ChemR23. Introduction of the Caenorhabditis elegans Fat1 transgene, leading to an endogenous omega-3 fatty acid synthesis and hence increased substrate for resolvin E1 formation, significantly diminished the differences in phosphate-induced calcification between ChemR23+/+ and ChemR23-/- mice. CONCLUSION: This study identifies ChemR23 as a previously unrecognized determinant of synthetic and osteoblastic smooth muscle cell phenotype, favouring phosphate-induced vascular calcification. This effect may be of particular importance in the absence of ChemR23 ligands, such as resolvin E1, which acts as a calcification inhibitor under hyperphosphatic conditions.


Assuntos
Adaptação Fisiológica , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteogênese , Fosfatos/metabolismo , Receptores de Quimiocinas/metabolismo , Calcificação Vascular/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Adulto , Idoso , Animais , Caderinas/genética , Caderinas/metabolismo , Colecalciferol , Modelos Animais de Doenças , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacologia , Feminino , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Osteogênese/efeitos dos fármacos , Ratos , Receptores de Quimiocinas/deficiência , Receptores de Quimiocinas/efeitos dos fármacos , Receptores de Quimiocinas/genética , Transdução de Sinais , Calcificação Vascular/induzido quimicamente , Calcificação Vascular/patologia , Calcificação Vascular/prevenção & controle
20.
Nephrol Dial Transplant ; 34(12): 2051-2057, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30312430

RESUMO

BACKGROUND: Recent studies suggest that the phosphaturic hormone fibroblast growth factor 23 (FGF23) is involved in regulation of renal sodium excretion and blood pressure. There is evidence of both direct effects via regulation of the sodium-chloride symporter (NCC) in the distal tubule, and indirect effects through interactions with the renin-angiotensin-aldosterone system. However, clinical data on the association between FGF23 and renal sodium regulation is lacking. Herein, we investigated the associations of FGF23 with renal sodium handling and blood pressure in non-dialysis CKD patients. METHODS: This was a cross-sectional study encompassing 180 CKD patients Stage 1-5, undergoing renal biopsy. Plasma intact FGF23, 24-h urinary sodium excretion, fractional excretion of sodium (FENa) and blood pressure were measured at baseline. The association between FGF23 and renal sodium handling was explored by multivariate regression analysis. RESULTS: The median age was 52.8 years, 60.6% were men and the median estimated glomerular filtration rate (eGFR) was 50.6 mL/min/1.73 m2. In univariate analysis, FGF23 was positively associated with FENa (Spearman's rho = 0.47; P < 0.001) and systolic blood pressure (rho = 0.17, P < 0.05), but not with plasma sodium, 24-h urinary sodium excretion or mean arterial blood pressure. The association between FGF23 and FENa remained significant after adjustment for potential confounders (multivariable adjusted ß coefficient 0.60, P < 0.001). This association was stronger among the 107 individuals with eGFR <60 mL/min/1.73 m2 (ß = 0.47, P = 0.04) and in the 73 individuals on any diuretics (ß = 0.88, P < 0.001). Adjustment for measured GFR instead of eGFR did not alter the relationship. CONCLUSIONS: FGF23 is independently associated with increased FENa in non-dialysis CKD patients. These data do not support the notion that FGF23 causes clinically significant sodium retention. Further studies are warranted to explore the mechanism underlying this association.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Insuficiência Renal Crônica/urina , Sódio/urina , Adulto , Idoso , Pressão Sanguínea , Estudos Transversais , Feminino , Fator de Crescimento de Fibroblastos 23 , Taxa de Filtração Glomerular , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/patologia , Sódio/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA