Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.236
Filtrar
1.
Int J Mol Sci ; 25(17)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39273465

RESUMEN

The number of patients with end-stage renal disease (ESRD) requiring hemodialysis is increasing worldwide. Although arteriovenous fistula (AVF) is the best and most important vascular access (VA) for hemodialysis, its primary maturation failure rate is as high as 60%, which seriously endangers the prognosis of hemodialysis patients. After AVF establishment, the venous outflow tract undergoes hemodynamic changes, which are translated into intracellular signaling pathway cascades, resulting in an outward and inward remodeling of the vessel wall. Outward remodeling refers to the thickening of the vessel wall and the dilation of the lumen to accommodate the high blood flow in the AVF, while inward remodeling is mainly characterized by intimal hyperplasia. More and more studies have shown that the two types of remodeling are closely related in the occurrence and development of, and jointly determining the final fate of, AVF. Therefore, it is essential to investigate the underlying mechanisms involved in outward and inward remodeling for identifying the key targets in alleviating AVF dysfunction. In this review, we summarize the current clinical diagnosis, monitoring, and treatment techniques for AVF dysfunction and discuss the possible pathological mechanisms related to improper outward and inward remodeling in AVF dysfunction, as well as summarize the similarities and differences between the two remodeling types in molecular mechanisms. Finally, the representative therapeutic targets of potential clinical values are summarized.


Asunto(s)
Fístula Arteriovenosa , Diálisis Renal , Humanos , Fístula Arteriovenosa/metabolismo , Fístula Arteriovenosa/terapia , Fístula Arteriovenosa/patología , Remodelación Vascular , Fallo Renal Crónico/terapia , Fallo Renal Crónico/patología , Fallo Renal Crónico/metabolismo , Derivación Arteriovenosa Quirúrgica/efectos adversos , Animales , Hemodinámica , Transducción de Señal , Terapia Molecular Dirigida
2.
Tissue Cell ; 90: 102527, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39181089

RESUMEN

Chronic kidney disease (CKD) and end-stage renal disease (ESRD) are prevalent and debilitating conditions with a significant impact on patients' quality of life. In this study, we conducted a comprehensive investigation into the histological characteristics of renal progenitor/stem cells (RPCs), renal mesenchymal stem-like cells, and endothelial progenitor cells (EPCs) in CKD and ESRD patients. Additionally, we performed a molecular docking analysis to explore potential drug-receptor interactions involving common medications prescribed to CKD patients. Our histological examination revealed a noteworthy increase in the number of CD24- and CD133-positive cells in CKD and ESRD patients, representing RPCs. These cells are implicated in kidney repair and regeneration, underscoring their potential role in CKD management. Moreover, we observed an elevation in the number of EPCs within the kidneys of CKD and ESRD patients, suggesting a protective role of EPCs in kidney preservation. The molecular docking analysis unveiled intriguing insights into potential drug interventions. Notably, digoxin exhibited the highest in-silico binding affinity to numerous receptors associated with the functions of RPCs, renal mesenchymal stem-like cells, and EPCs, emphasizing the potential multifaceted effects of this cardiac glycoside in CKD patients. Other drugs, including apixaban, glimepiride, and glibenclamide, also displayed strong in-silico affinities to specific receptors, indicating their potential influence on various renal cell functions. In conclusion, this study provides valuable insights into the histological alterations in renal cell populations in CKD and ESRD patients and underscores the potential roles of RPCs and EPCs in kidney repair and preservation. The molecular docking analysis reveals the complex interactions between common drugs and renal cells, suggesting the need for further in-vitro and in-vivo research to fully understand these relationships. These findings contribute to our understanding of CKD and offer new avenues for research into potential therapeutic interventions.


Asunto(s)
Células Progenitoras Endoteliales , Fallo Renal Crónico , Células Madre Mesenquimatosas , Simulación del Acoplamiento Molecular , Insuficiencia Renal Crónica , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/metabolismo , Fallo Renal Crónico/patología , Fallo Renal Crónico/metabolismo , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/patología , Riñón/patología , Riñón/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Anciano , Adulto
3.
Int J Mol Sci ; 25(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39201577

RESUMEN

Exogenous and endogenous advanced glycation end products (AGEs) contribute to the pathogenesis and progression of renal disease. This is a one-month controlled dietary counseling trial that restricts nutritional AGEs in patients with end-stage renal disease (ESRD) undergoing haemodialysis (n = 22 participants in the intervention and n = 20 participants in the control group). Haematological, biochemical markers, the soluble form of the receptor for AGEs (sRAGE), and carboxymethyl lysine (CML) were measured at baseline and at follow-up. Mononuclear cells were isolated and the protein expression of RAGE and the inflammatory marker COX-2 was measured using Western immunoblotting. The intervention group presented a lower increase in CML compared to the control group (12.39% median change in the intervention vs. 69.34% in the control group, p = 0.013), while RAGE (% mean change -56.54 in the intervention vs. 46.51 in the control group, p < 0.001) and COX-2 (% mean change -37.76 in the intervention vs. 0.27 in the control group, p < 0.001) were reduced compared to the control group. sRAGE was reduced in both groups. In addition, HbA1c (at two months), total cholesterol, and triglycerides were reduced in the intervention versus the control group. The adoption of healthy cooking methods deserves further research as a possible way of modulating inflammatory markers in patients with CKD.


Asunto(s)
Productos Finales de Glicación Avanzada , Fallo Renal Crónico , Lisina , Receptor para Productos Finales de Glicación Avanzada , Diálisis Renal , Humanos , Productos Finales de Glicación Avanzada/metabolismo , Masculino , Femenino , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/dietoterapia , Persona de Mediana Edad , Lisina/análogos & derivados , Lisina/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Anciano , Inflamación/metabolismo , Biomarcadores , Adulto , Ciclooxigenasa 2/metabolismo
4.
BMC Nephrol ; 25(1): 268, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39179976

RESUMEN

BACKGROUND: Urinary Dickkopf 3 (DKK3) excretion is a recently established biomarker of renal functional development. Its excretion into the peritoneal cavity has not been reported. We here studied DKK3 in peritoneal dialysis. METHODS: DKK3 was assessed in serum, urine and dialysate in a prevalent adult peritoneal dialysis cohort and its concentration analyzed in relation to creatinine and clinical characteristics. RESULTS: Highest DKK3 concentrations were found in serum, followed by urine. Dialysate concentrations were significantly lower. Dialysate DKK3 correlated with both other compartments. Serum, dialysate and urine values were stable during three months of follow-up. Continuous ambulatory dialysis (CAPD) but not cycler-assisted peritoneal dialysis (CCPD) volume-dependently increased peritoneal DKK3 in relation to creatinine. RAAS blockade significantly decreased urinary, but not serum or peritoneal DKK3. CONCLUSION: Our data provide a detailed characterization of DKK3 in peritoneal dialysis. They support the notion that the RAAS system is essential for renal DKK3 handling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Diálisis Peritoneal , Humanos , Masculino , Femenino , Persona de Mediana Edad , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quimiocinas/sangre , Quimiocinas/metabolismo , Anciano , Adulto , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Biomarcadores/sangre , Soluciones para Diálisis/metabolismo , Riñón/metabolismo , Peritoneo/metabolismo , Diálisis Peritoneal Ambulatoria Continua , Sistema Renina-Angiotensina/fisiología , Creatinina/orina , Creatinina/sangre , Creatinina/metabolismo
5.
BMC Nephrol ; 25(1): 227, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39020292

RESUMEN

BACKGROUND: End-stage and acquired cystic renal disease (ESRD/ACRD) kidneys are characterized by inflammatory remodelling and multiplex renal cell carcinomas (RCC). Eosinophilic vacuolated tumour (EVT) occurs exclusively in ACRD. The aim of this study was to identify the involvement of thioredoxin-interacting protein (TXNIP) and thioredoxin (TXN) in ESRD/ACRD pathology. METHODS: Expression of TXNIP and TXN was examined in histological slides of 6 ESRD and 6 ACRD kidneys, precursor lesions and associated tumours as well as of RCCs from the general population by immunohistochemistry. RESULTS: Strong TXNIP expression was seen in epithelial cells, myo-fibroblasts and endothelial cells and weak TXN expression in ESRD/ACRD kidneys and tumours. In ACRD specific EVT and its precursors TXN were translocated into nuclei. CONCLUSION: The impaired TXNIP/TXN redox homeostasis might be associated with development of multiplex cancer especially of EVT in ESRD/ACRD kidney.


Asunto(s)
Proteínas Portadoras , Fallo Renal Crónico , Neoplasias Renales , Tiorredoxinas , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Proteínas Portadoras/metabolismo , Tiorredoxinas/metabolismo , Fallo Renal Crónico/metabolismo , Masculino , Persona de Mediana Edad , Femenino , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Anciano , Núcleo Celular/metabolismo , Adulto , Transporte Activo de Núcleo Celular
6.
Elife ; 122024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980302

RESUMEN

Trained immunity is the long-term functional reprogramming of innate immune cells, which results in altered responses toward a secondary challenge. Despite indoxyl sulfate (IS) being a potent stimulus associated with chronic kidney disease (CKD)-related inflammation, its impact on trained immunity has not been explored. Here, we demonstrate that IS induces trained immunity in monocytes via epigenetic and metabolic reprogramming, resulting in augmented cytokine production. Mechanistically, the aryl hydrocarbon receptor (AhR) contributes to IS-trained immunity by enhancing the expression of arachidonic acid (AA) metabolism-related genes such as arachidonate 5-lipoxygenase (ALOX5) and ALOX5 activating protein (ALOX5AP). Inhibition of AhR during IS training suppresses the induction of IS-trained immunity. Monocytes from end-stage renal disease (ESRD) patients have increased ALOX5 expression and after 6 days training, they exhibit enhanced TNF-α and IL-6 production to lipopolysaccharide (LPS). Furthermore, healthy control-derived monocytes trained with uremic sera from ESRD patients exhibit increased production of TNF-α and IL-6. Consistently, IS-trained mice and their splenic myeloid cells had increased production of TNF-α after in vivo and ex vivo LPS stimulation compared to that of control mice. These results provide insight into the role of IS in the induction of trained immunity, which is critical during inflammatory immune responses in CKD patients.


Asunto(s)
Indicán , Fallo Renal Crónico , Receptores de Hidrocarburo de Aril , Animales , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/genética , Fallo Renal Crónico/inmunología , Fallo Renal Crónico/metabolismo , Humanos , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Ácido Araquidónico/metabolismo , Masculino , Inmunidad Innata/efectos de los fármacos , Ratones Endogámicos C57BL , Araquidonato 5-Lipooxigenasa/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Inmunidad Entrenada
7.
Biomolecules ; 14(6)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38927027

RESUMEN

Microbiota tryptophan metabolism and the biosynthesis of indole derivatives play an important role in homeostasis and pathogenesis in the human body and can be affected by the gut microbiota. However, studies on the interplay between gut microbiota and tryptophan metabolites in patients undergoing dialysis are lacking. This study aimed to identify the gut microbiota, the indole pathway in tryptophan metabolism, and significant functional differences in ESRD patients with regular hemodialysis. We performed the shotgun metagenome sequencing of stool samples from 85 hemodialysis patients. Using the linear discriminant analysis effect size (LEfSe), we examined the composition of the gut microbiota and metabolic features across varying concentrations of tryptophan and indole metabolites. Higher tryptophan levels promoted tyrosine degradation I and pectin degradation I metabolic modules; lower tryptophan levels were associated with glutamate degradation I, fructose degradation, and valine degradation modules. Higher 3-indoxyl sulfate concentrations were characterized by alanine degradation I, anaerobic fatty acid beta-oxidation, sulfate reduction, and acetyl-CoA to crotonyl-CoA. Contrarily, lower 3-indoxyl sulfate levels were related to propionate production III, arabinoxylan degradation, the Entner-Doudoroff pathway, and glutamate degradation II. The present study provides a better understanding of the interaction between tryptophan, indole metabolites, and the gut microbiota as well as their gut metabolic modules in ESRD patients with regular hemodialysis.


Asunto(s)
Microbioma Gastrointestinal , Indoles , Diálisis Renal , Triptófano , Humanos , Triptófano/metabolismo , Indoles/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Anciano , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/microbiología , Heces/microbiología , Redes y Vías Metabólicas , Adulto , Metagenoma
8.
BMC Nephrol ; 25(1): 155, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38702607

RESUMEN

BACKGROUND: Oxidative stress, an imbalance between reactive oxygen species production and antioxidant capacity, increases in patients with coronavirus disease (COVID-19) or renal impairment. We investigated whether combined COVID-19 and end-stage renal disease (ESRD) would increase oxidative stress levels compared to each disease alone. METHODS: Oxidative stress was compared among three groups. Two groups comprised patients with COVID-19 referred to the hospital with or without renal impairment (COVID-ESRD group [n = 18]; COVID group [n = 17]). The third group (ESRD group [n = 18]) comprised patients without COVID-19 on maintenance hemodialysis at a hospital. RESULTS: The total oxidative stress in the COVID-ESRD group was lower than in the COVID group (p = 0.047). The total antioxidant status was higher in the COVID-ESRD group than in the ESRD (p < 0.001) and COVID (p < 0.001) groups after controlling for covariates. The oxidative stress index was lower in the COVID-ESRD group than in the ESRD (p = 0.001) and COVID (p < 0.001) groups. However, the three oxidative parameters did not differ significantly between the COVID and COVID-ESRD groups. CONCLUSIONS: The role of reactive oxygen species in the pathophysiology of COVID-19 among patients withESRD appears to be non-critical. Therefore, the provision of supplemental antioxidants may not confer a therapeutic advantage, particularly in cases of mild COVID-19 in ESRD patients receiving hemodialysis. Nonetheless, this area merits further research.


Asunto(s)
COVID-19 , Fallo Renal Crónico , Estrés Oxidativo , Humanos , COVID-19/complicaciones , COVID-19/metabolismo , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/complicaciones , Proyectos Piloto , Masculino , Femenino , Persona de Mediana Edad , Anciano , Antioxidantes/metabolismo , Diálisis Renal , SARS-CoV-2 , Especies Reactivas de Oxígeno/metabolismo
9.
Int J Mol Sci ; 25(9)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38731843

RESUMEN

Chronic kidney disease (CKD) is a global health concern affecting approximately one billion individuals worldwide. End-stage kidney disease (ESKD), the most severe form of CKD, is often accompanied by anemia. Peritoneal dialysis (PD), a common treatment for ESKD, utilizes the peritoneum for solute transfer but is associated with complications including protein loss, including transferrin (Tf) a key protein involved in iron transport. This study investigated Tf characteristics in ESKD patients compared to healthy individuals using lectin microarray, spectroscopic techniques and immunocytochemical analysis to assess Tf interaction with transferrin receptors (TfRs). ESKD patients exhibited altered Tf glycosylation patterns, evidenced by significant changes in lectin reactivity compared to healthy controls. However, structural analyses revealed no significant differences in the Tf secondary or tertiary structures between the two groups. A functional analysis demonstrated comparable Tf-TfR interaction in both PD and healthy samples. Despite significant alterations in Tf glycosylation, structural integrity and Tf-TfR interaction remained preserved in PD patients. These findings suggest that while glycosylation changes may influence iron metabolism, they do not impair Tf function. The study highlights the importance of a glucose-free dialysis solutions in managing anemia exacerbation in PD patients with poorly controlled anemia, potentially offering a targeted therapeutic approach to improve patient outcomes.


Asunto(s)
Fallo Renal Crónico , Receptores de Transferrina , Transferrina , Humanos , Transferrina/metabolismo , Glicosilación , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Receptores de Transferrina/metabolismo , Diálisis Peritoneal , Anciano , Adulto , Hierro/metabolismo
10.
Semin Dial ; 37(3): 259-268, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38506151

RESUMEN

BACKGROUND: Dialytic phosphate removal is a cornerstone of the management of hyperphosphatemia in peritoneal dialysis (PD) patients, but the influencing factors on peritoneal phosphate clearance (PPC) are incompletely understood. Our objective was to explore clinically relevant factors associated with PPC in patients with different PD modality and peritoneal transport status and the association of PPC with mortality. METHODS: This is a cross-sectional and prospective observational study. Four hundred eighty-five PD patients were enrolled and divided into 2 groups according to PPC. All-cause mortality was evaluated after followed-up for at least 3 months. RESULTS: High PPC group showed lower mortality compared with Low PPC group by Kaplan-Meier analysis and log-rank test. Both multivariate linear regression and multivariate logistic regression revealed that high transport status, total effluent dialysate volume per day, continuous ambulatory PD (CAPD), and protein in total effluent dialysate volume appeared to be positively correlated with PPC; body mass index (BMI) and the normalized protein equivalent of total nitrogen appearance (nPNA) were negatively correlated with PPC. Besides PD modality and membrane transport status, total effluent dialysate volume showed a strong relationship with PPC, but the correlation differed among PD modalities. CONCLUSIONS: Higher PPC was associated with lower all-cause mortality risk in PD patients. Higher PPC correlated with CAPD modality, fast transport status, higher effluent dialysate volume and protein content, and with lower BMI and nPNA.


Asunto(s)
Hiperfosfatemia , Fallo Renal Crónico , Diálisis Peritoneal , Fosfatos , Humanos , Masculino , Femenino , Persona de Mediana Edad , Estudios Prospectivos , Diálisis Peritoneal/mortalidad , Estudios Transversales , Fosfatos/metabolismo , Fosfatos/análisis , Hiperfosfatemia/etiología , Fallo Renal Crónico/terapia , Fallo Renal Crónico/mortalidad , Fallo Renal Crónico/metabolismo , Anciano , Diálisis Peritoneal Ambulatoria Continua/mortalidad , Soluciones para Diálisis , Adulto
11.
J Ren Nutr ; 34(5): 463-468, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38490516

RESUMEN

OBJECTIVES: Low plasma folate levels have been reported in patients undergoing hemodialysis and peritoneal dialysis (PD) in clinical studies. However, folate transport has never been mentioned as a factor contributing to low plasma folate levels in patients undergoing PD. The peritoneal equilibrium test (PET) assesses the plasma creatinine level and glucose transport abilities. This study aimed to evaluate the association between plasma folate levels and folate transport during PD based on PET grades. METHODS: This study recruited 50 patients who underwent PD for ≥3 months and were categorized according to PET grades. Data regarding plasma folate levels and dialysate folate were collected. The primary outcomes were the relationship between the PET grade and plasma folate level and between the PET grade and dialysate-to-plasma folate concentration ratio (D/P folate). Furthermore, the difference in the plasma folate level and D/P folate between men and women was assessed. RESULTS: The plasma folate level and the D/P folate significantly differed among the 4 PET groups (both P < .001). PET grade was significantly negatively correlated with plasma folate levels (r = -0.56, P < .001) and positively correlated with D/P folate (r = 0.686, P < .001). In subgroup analysis, neither the plasma folate level nor the D/P folate significantly differed between men and women. CONCLUSIONS: Our study provides clinical evidence that the PET grade is associated with the plasma folate level and D/P folate, regardless of sex. Larger cohort studies are warranted to assess the importance of folate supplementation during PD based on PET grades.


Asunto(s)
Ácido Fólico , Diálisis Peritoneal , Humanos , Ácido Fólico/sangre , Femenino , Masculino , Persona de Mediana Edad , Diálisis Peritoneal/métodos , Anciano , Transporte Biológico , Peritoneo/metabolismo , Creatinina/sangre , Creatinina/metabolismo , Adulto , Soluciones para Diálisis/metabolismo , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo
12.
Amyloid ; 31(2): 105-115, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38343068

RESUMEN

BACKGROUND: Dialysis-related amyloidosis (DRA) is a severe complication in end-stage kidney disease (ESKD) patients undergoing long-term dialysis treatment, characterized by the deposition of ß2-microglobulin-related amyloids (Aß2M amyloid). To inhibit DRA progression, hexadecyl-immobilized cellulose bead (HICB) columns are employed to adsorb circulating ß2-microglobulin (ß2M). However, it is possible that the HICB also adsorbs other molecules involved in amyloidogenesis. METHODS: We enrolled 14 ESKD patients using HICB columns for DRA treatment; proteins were extracted from HICBs following treatment and identified using liquid chromatography-linked mass spectrometry. We measured the removal rate of these proteins and examined the effect of those molecules on Aß2M amyloid fibril formation in vitro. RESULTS: We identified 200 proteins adsorbed by HICBs. Of these, 21 were also detected in the amyloid deposits in the carpal tunnels of patients with DRA. After passing through the HICB column and hemodialyzer, the serum levels of proteins such as ß2M, lysozyme, angiogenin, complement factor D and matrix Gla protein were reduced. These proteins acted in the Aß2M amyloid fibril formation. CONCLUSIONS: HICBs adsorbed diverse proteins in ESKD patients with DRA, including those detected in amyloid lesions. Direct hemoperfusion utilizing HICBs may play a role in acting Aß2M amyloidogenesis by reducing the amyloid-related proteins.


Asunto(s)
Amiloidosis , Celulosa , Fallo Renal Crónico , Proteómica , Diálisis Renal , Microglobulina beta-2 , Humanos , Amiloidosis/metabolismo , Amiloidosis/sangre , Amiloidosis/terapia , Diálisis Renal/efectos adversos , Masculino , Femenino , Microglobulina beta-2/metabolismo , Microglobulina beta-2/sangre , Proteómica/métodos , Anciano , Celulosa/química , Persona de Mediana Edad , Adsorción , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/sangre , Espectrometría de Masas/métodos , Amiloide/metabolismo , Cromatografía Liquida
13.
Pediatr Nephrol ; 39(5): 1521-1532, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38051389

RESUMEN

BACKGROUND: Hemodialysis is a life-saving technology used during periods of acute or chronic kidney failure to remove toxins, and maintain fluid, electrolyte and metabolic balance. While this technology plays an important role for pediatric patients with kidney dysfunction, it can alter the pharmacokinetic behavior of medications placing patients at risk for suboptimal dosing and drug toxicity. The ability to directly translate pharmacokinetic alterations into dosing recommendations has thus far been limited and dosing guidance specific to pediatric hemodialysis patients is rare. Despite differences in dialysis prescription and patient populations, intermittent (iHD) and continuous kidney replacement therapy (CKRT) patients are often pooled together. In order to develop evidence-based dosing guidelines, it is important to first prioritize drugs for study in each modality. METHODS: Here we aim to identify priority drugs in two hemodialysis modalities, through: 1) Identification of hospitalized, pediatric patients who received CKRT or intermittent hemodialysis (iHD) using a machine learning-based predictive model based on medications; 2) Identification of medication administration patterns in these patient cohorts; and 3) Identification of the most commonly prescribed drugs that lack published dosing guidance. RESULTS: Notable differences were found in the pattern of medications and drug dosing guidance between iHD and CKRT patients. Antibiotics, diuretics and sedatives were more common in CKRT patients. Out of the 50 most commonly administered medications in the two modalities, only 34% and 28% had dosing guidance present for iHD and CKRT, respectively. CONCLUSIONS: Our results add to the understanding of the differences between iHD and CKRT patient populations by identifying commonly used medications that lack dosing guidance for each hemodialysis modality, helping to pinpoint priority medications for further study. Overall, this study provides an overview of the current limitations in medication use in this at-risk population, and provides a framework for future studies by identifying commonly used medications in pediatric CKRT and iHD patients.


Asunto(s)
Lesión Renal Aguda , Terapia de Reemplazo Renal Continuo , Fallo Renal Crónico , Niño , Humanos , Lesión Renal Aguda/epidemiología , Antibacterianos/uso terapéutico , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Preparaciones Farmacéuticas , Diálisis Renal/métodos , Terapia de Reemplazo Renal
14.
Clin Exp Nephrol ; 28(3): 181-191, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37882850

RESUMEN

INTRODUCTION: Diabetic kidney disease (DKD) is one of the prominent microvascular complications of diabetes and the leading cause of end-stage renal disease. Inflammation plays a crucial role in the development and progression of DKD. Currently, only a few studies depict the landscape of infiltrating immune cells and their potential regulatory network in DKD. To gain a better understanding of the role of immune cells in the renal microenvironment, we sought to reveal the profile of infiltrating immune cells and their potential regulatory network in DKD. METHODS: We obtained the transcriptomes and the corresponding clinical data of 19 DKD and 25 control samples from the Gene Expression Omnibus and Nephroseq databases, respectively. Thereafter, we conducted an analysis on the infiltrating immune cells and identified immune-related differentially expressed genes through bioinformatics. Finally, correlation analyses among immune cells, immune genes, and clinical manifestations were performed, and differentially infiltrating immune cell subsets were verified through multiplex immunofluorescence staining. RESULTS: We demonstrated the landscape of infiltrating immune cells in patients with DKD and identified the top five hub immune regulatory genes (C3, IL7R, TYROBP, BMP2, and CXCL6). Three of the core genes (C3, BMP2, and CXCL6) were significantly correlated with the estimated glomerular filtration rate. Through multiplex immunofluorescence staining, we verified that macrophage numbers were remarkably elevated, whereas Treg cells were remarkably reduced in diabetic kidney tissues. Th2 cells were scarce in the kidney tissue. CONCLUSION: Collectively, our findings shed light on new, possible therapeutic strategies for DKD, from an immune microenvironment perspective.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Fallo Renal Crónico , Humanos , Riñón , Fallo Renal Crónico/metabolismo , Biología Computacional , Tasa de Filtración Glomerular
15.
Exp Anim ; 73(1): 29-40, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-37482420

RESUMEN

Diabetic nephropathy (DN), included in diabetic kidney disease (DKD), is a primary driver of end-stage renal disease (ESRD) leading to dialysis treatment. To develop new therapeutic drugs to prevent ESRD and avoid dialysis treatment, insight into DKD pathophysiology and animal models suitable for drug efficacy testing are needed. In this study, transcriptome analysis of kidneys from 26-week-old and 35-week-old uninephrectomized (UNX) db/db mice was used to identify the pathways that affect the deterioration of renal function in db/db mice. Differentially expressed genes suggested that there was increased interferon (IFN)-γ signaling during the 26 to 35-week period. Modules that changed between 26 and 35 weeks of age extracted by weighted gene co-expression network analysis (WGCNA) suggested increased the tumor necrosis factor (TNF)-α and nuclear factor-kappa B (NF-κB) signaling pathway in component cells of glomeruli. The protein-protein interaction (PPI) network analysis identified Cxcl16 as a hub gene for those signaling pathways, and it was shown that the pathways in this module changed when the glomerular filtration rate decreased in patients with DN. These results suggested the possibility that signaling mediated by Cxcl16 induced by IFN-γ and TNF-α between 26 and 35 weeks of age leads to renal fibrosis, resulting in severe disease. Drugs that target such pathways can be options for developing drugs for DN. We also think that the uninephrectomized db/db mouse can be used as an animal model of severe DKD and to evaluate efficacy in patients with DN.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Fallo Renal Crónico , Ratones , Humanos , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/tratamiento farmacológico , Riñón , Transducción de Señal/genética , Ratones Endogámicos , Factor de Necrosis Tumoral alfa/metabolismo , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/patología , Perfilación de la Expresión Génica
16.
Pharmacol Res ; 198: 107009, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37995896

RESUMEN

Although hyperhomocysteinemia (hHcys) has been recognized as an important independent risk factor in the progression of end-stage renal disease and the development of cardiovascular complications related to end-stage renal disease, the mechanisms triggering pathogenic actions of hHcys are not fully understood. The present study was mainly designed to investigate the role of HDACs in renal injury induced by hHcys. Firstly, we identified the expression patterns of HDACs and found that, among zinc-dependent HDACs, HDAC9 was preferentially upregulated in the kidney from mice with hHcys. Deficiency or pharmacological inhibition of HDAC9 ameliorated renal injury in mice with hHcys. Moreover, podocyte-specific deletion of HDAC9 significantly attenuated podocyte injury and proteinuria. In vitro, gene silencing of HDAC9 attenuated podocyte injury by inhibiting apoptosis, reducing oxidative stress and maintaining the expressions of podocyte slit diaphragm proteins. Mechanically, we proved for the first time that HDAC9 reduced the acetylation level of H3K9 in the promoter of Klotho, then inhibited gene transcription of Klotho, finally aggravating podocyte injury in hHcys. In conclusion, our results indicated that targeting of HDAC9 might be an attractive therapeutic strategy for the treatment of renal injury induced by hHcys.


Asunto(s)
Hiperhomocisteinemia , Fallo Renal Crónico , Podocitos , Animales , Ratones , Represión Epigenética , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/genética , Fallo Renal Crónico/metabolismo , Podocitos/patología
17.
Curr Drug Metab ; 24(10): 684-699, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37927072

RESUMEN

AIMS: To identify single nucleotide polymorphisms (SNPs) of paracetamol-metabolizing enzymes that can predict acute liver injury. BACKGROUND: Paracetamol is a commonly administered analgesic/antipyretic in critically ill and chronic renal failure patients and several SNPs influence the therapeutic and toxic effects. OBJECTIVE: To evaluate the role of machine learning algorithms (MLAs) and bioinformatics tools to delineate the predictor SNPs as well as to understand their molecular dynamics. METHODS: A cross-sectional study was undertaken by recruiting critically ill patients with chronic renal failure and administering intravenous paracetamol as a standard of care. Serum concentrations of paracetamol and the principal metabolites were estimated. Following SNPs were evaluated: CYP2E1*2, CYP2E1_-1295G>C, CYP2D6*10, CYP3A4*1B, CYP3A4*2, CYP1A2*1K, CYP1A2*6, CYP3A4*3, and CYP3A5*7. MLAs were used to identify the predictor genetic variable for acute liver failure. Bioinformatics tools such as Predict SNP2 and molecular docking (MD) were undertaken to evaluate the impact of the above SNPs with binding affinity to paracetamol. RESULTS: CYP2E1*2 and CYP1A2*1C genotypes were identified by MLAs to significantly predict hepatotoxicity. The predictSNP2 revealed that CYP1A2*3 was highly deleterious in all the tools. MD revealed binding energy of -5.5 Kcal/mol, -6.9 Kcal/mol, and -6.8 Kcal/mol for CYP1A2, CYP1A2*3, and CYP1A2*6 against paracetamol. MD simulations revealed that CYP1A2*3 and CYP1A2*6 missense variants in CYP1A2 affect the binding ability with paracetamol. In-silico techniques found that CYP1A2*2 and CYP1A2*6 are highly harmful. MD simulations revealed CYP3A4*2 (A>G) had decreased binding energy with paracetamol than CYP3A4, and CYP3A4*2(A>T) and CYP3A4*3 both have greater binding energy with paracetamol. CONCLUSION: Polymorphisms in CYP2E1, CYP1A2, CYP3A4, and CYP3A5 significantly influence paracetamol's clinical outcomes or binding affinity. Robust clinical studies are needed to identify these polymorphisms' clinical impact on the pharmacokinetics or pharmacodynamics of paracetamol.


Asunto(s)
Citocromo P-450 CYP1A2 , Fallo Renal Crónico , Humanos , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Citocromo P-450 CYP2E1/genética , Citocromo P-450 CYP2E1/metabolismo , Acetaminofén/efectos adversos , Acetaminofén/metabolismo , Polimorfismo de Nucleótido Simple , Simulación del Acoplamiento Molecular , Enfermedad Crítica , Estudios Transversales , Hígado/metabolismo , Fallo Renal Crónico/metabolismo , Aprendizaje Automático Supervisado , Algoritmos
18.
J Clin Invest ; 133(20)2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37843281

RESUMEN

Enabling the early detection and prevention of diabetic kidney damage has potential to substantially reduce the global burden of kidney failure. There is a critical need for identification of mechanistic biomarkers that can predict progression and serve as therapeutic targets. In this issue of the JCI, Sharma and colleagues used an integrated multiomics approach to identify the metabolite adenine as a noninvasive biomarker of progression in early diabetic kidney disease (DKD). The highest tertile of urine adenine/creatinine ratio (UAdCR) was associated with higher risk for end-stage kidney disease and mortality across independent cohorts, including participants with early DKD without macroalbuminuria. Spatial metabolomics, single-cell transcriptomics, and experimental studies localized adenine to regions of tubular pathology and implicated the mTOR pathway in adenine-mediated tissue fibrosis. Inhibition of endogenous adenine production was protective in a diabetic model. These findings exemplify the potential for multiomics to uncover mechanistic biomarkers and targeted therapies in DKD.


Asunto(s)
Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Fallo Renal Crónico , Humanos , Nefropatías Diabéticas/diagnóstico , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Adenina , Fallo Renal Crónico/metabolismo , Biomarcadores/metabolismo , Metabolómica , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Riñón/metabolismo
19.
Int J Mol Sci ; 24(15)2023 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-37569805

RESUMEN

Patients with end-stage kidney disease (ESKD) suffer from high levels of protein-bound uremic toxins (PBUTs) that contribute to various comorbidities. Conventional dialysis methods are ineffective in removing these PBUTs. A potential solution could be offered by a bioartificial kidney (BAK) composed of porous membranes covered by proximal tubule epithelial cells (PTECs) that actively secrete PBUTs. However, BAK development is currently being hampered by a lack of knowledge regarding the cytocompatibility of the dialysis fluid (DF) that comes in contact with the PTECs. Here, we conducted a comprehensive functional assessment of the DF on human conditionally immortalized PTECs (ciPTECs) cultured as monolayers in well plates, on Transwell® inserts, or on hollow fiber membranes (HFMs) that form functional units of a BAK. We evaluated cell viability markers, monolayer integrity, and PBUT clearance. Our results show that exposure to DF did not affect ciPTECs' viability, membrane integrity, or function. Seven anionic PBUTs were efficiently cleared from the perfusion fluid containing a PBUTs cocktail or uremic plasma, an effect which was enhanced in the presence of albumin. Overall, our findings support that the DF is cytocompatible and does not compromise ciPTECs function, paving the way for further advancements in BAK development and its potential clinical application.


Asunto(s)
Fallo Renal Crónico , Toxinas Biológicas , Humanos , Diálisis Renal/métodos , Tóxinas Urémicas , Fallo Renal Crónico/terapia , Fallo Renal Crónico/metabolismo , Riñón/metabolismo , Túbulos Renales Proximales/metabolismo , Soluciones para Diálisis/metabolismo , Toxinas Biológicas/metabolismo
20.
J Nephrol ; 36(6): 1673-1681, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37439962

RESUMEN

BACKGROUND: End-stage kidney disease and acquired cystic kidney disease are the final stages of chronic kidney disease, leading to loss of kidney function and frequent development of tumours. It has been suggested that an inflammatory microenvironment may be responsible for the progressive kidney remodelling and cancer development. METHODS: Our aim was to analyse gene expression suggested to be involved in the remodelling of kidneys in end-stage kidney disease, and in the development of preneoplastic lesions and tumours. Immunohistochemistry was employed to assess the cellular localisation of different genes involved in these pathways on representative tissue sections. RESULTS: Cellular (αSMA positive naïve activated fibroblasts, endothelial cells, macrophages) and non-cellular components (cytokines IL6, TGFß, IL1ß, CSF2, fibronectin, laminin, and matrix modifier proteases MMP9 and MMP12) of the inflammatory microenvironment were expressed in the kidneys of patients with end-stage kidney disease. IL6 and FN1 expressing naïve activated fibroblasts and recruited inflammatory cells were the most abundant cellular components of the inflammatory microenvironment. CONCLUSION: The progressive inflammatory and fibrotic processes in end-stage kidney disease have features recalling those of  a never healing wound and may explain the frequent development of kidney cancer.


Asunto(s)
Carcinoma de Células Renales , Fallo Renal Crónico , Neoplasias Renales , Humanos , Interleucina-6 , Células Endoteliales/patología , Neoplasias Renales/patología , Fallo Renal Crónico/metabolismo , Riñón/patología , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA