Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.410
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 42(1): 207-233, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38211945

RESUMEN

The immune system and the kidneys are closely related. Immune components mediate acute kidney disease and are crucial to the progression of chronic kidney disease. Beyond its pathogenic functions, the immune system supports immunological homeostasis in healthy kidneys. The kidneys help maintain immune equilibrium by removing metabolic waste products and toxins, thereby limiting local and systemic inflammation. In this review, we describe the close relationship between the immune system and the kidneys. We discuss how the imbalance in the immune response can be deleterious to the kidneys and how immunomodulation can be important in preventing end-stage renal disease. In addition, recent tools such as in silico platforms and kidney organoids can help unveil the relationship between immune cells and kidney homeostasis.


Asunto(s)
Enfermedades Renales , Humanos , Animales , Enfermedades Renales/inmunología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Riñón/inmunología , Riñón/metabolismo , Homeostasis , Inmunomodulación , Susceptibilidad a Enfermedades
2.
CA Cancer J Clin ; 71(1): 47-77, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32853404

RESUMEN

Onconephrology is a new subspecialty of nephrology that recognizes the important intersections of kidney disease with cancer. This intersection takes many forms and includes drug-induced nephrotoxicity, electrolyte disorders, paraneoplastic glomerulonephritis, and the interactions of chronic kidney disease with cancer. Data clearly demonstrate that, when patients with cancer develop acute or chronic kidney disease, outcomes are inferior, and the promise of curative therapeutic regimens is lessened. This highlights the imperative for collaborative care between oncologists and nephrologists in recognizing and treating kidney disease in patients with cancer. In response to this need, specific training programs in onconephrology as well as dedicated onconephrology clinics have appeared. This comprehensive review covers many of the critical topics in onconephrology, with a focus on acute kidney injury, chronic kidney disease, drug-induced nephrotoxicity, kidney disease in stem cell transplantation, and electrolyte disorders in patients with cancer.


Asunto(s)
Enfermedades Renales/terapia , Oncología Médica/métodos , Neoplasias/terapia , Nefrología/métodos , Antineoplásicos/efectos adversos , Humanos , Comunicación Interdisciplinaria , Enfermedades Renales/diagnóstico , Enfermedades Renales/etiología , Neoplasias/complicaciones , Neoplasias/diagnóstico , Trasplante de Células Madre/efectos adversos
3.
Proc Natl Acad Sci U S A ; 121(42): e2405860121, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39392665

RESUMEN

Tumors can induce systemic disturbances in distant organs, leading to physiological changes that enhance host morbidity. In Drosophila cancer models, tumors have been known for decades to cause hypervolemic "bloating" of the abdominal cavity. Here we use allograft and transgenic tumors to show that hosts display fluid retention associated with autonomously defective secretory capacity of fly renal tubules, which function analogous to those of the human kidney. Excretion from these organs is blocked by abnormal cells that originate from inappropriate activation of normally quiescent renal stem cells (RSCs). Blockage is initiated by IL-6-like oncokines that perturb renal water-transporting cells and trigger a damage response in RSCs that proceeds pathologically. Thus, a chronic inflammatory state produced by the tumor causes paraneoplastic fluid dysregulation by altering cellular homeostasis of host renal units.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre , Animales , Células Madre/metabolismo , Inflamación/patología , Humanos , Túbulos Renales/patología , Túbulos Renales/metabolismo , Drosophila melanogaster , Enfermedades Renales/patología , Enfermedades Renales/etiología , Enfermedades Renales/inmunología , Riñón/patología , Riñón/metabolismo , Síndromes Paraneoplásicos/inmunología , Síndromes Paraneoplásicos/metabolismo , Síndromes Paraneoplásicos/patología , Animales Modificados Genéticamente , Interleucina-6/metabolismo , Drosophila
4.
Circulation ; 150(16): e280-e295, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39253806

RESUMEN

Early identification of kidney dysfunction in patients with advanced heart failure is crucial for timely interventions. In addition to elevations in serum creatinine, kidney dysfunction encompasses inadequate maintenance of sodium and volume homeostasis, retention of uremic solutes, and disrupted endocrine functions. Hemodynamic derangements and maladaptive neurohormonal upregulations contribute to fluctuations in kidney indices and electrolytes that may recover with guideline-directed medical therapy. Quantifying the extent of underlying irreversible intrinsic kidney disease is crucial in predicting whether optimization of congestion and guideline-directed medical therapy can stabilize kidney function. This scientific statement focuses on clinical management of patients experiencing kidney dysfunction through the trajectory of advanced heart failure, with specific focus on (1) the conceptual framework for appropriate evaluation of kidney dysfunction within the context of clinical trajectories in advanced heart failure, including in the consideration of advanced heart failure therapies; (2) preoperative, perioperative, and postoperative approaches to evaluation and management of kidney disease for advanced surgical therapies (durable left ventricular assist device/heart transplantation) and kidney replacement therapies; and (3) the key concepts in palliative care and decision-making processes unique to individuals with concomitant advanced heart failure and kidney disease.


Asunto(s)
Insuficiencia Cardíaca , Enfermedades Renales , Humanos , American Heart Association , Insuficiencia Cardíaca/terapia , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/complicaciones , Corazón Auxiliar , Riñón/fisiopatología , Enfermedades Renales/diagnóstico , Enfermedades Renales/etiología , Enfermedades Renales/fisiopatología , Enfermedades Renales/terapia , Cuidados Paliativos/métodos , Estados Unidos/epidemiología , Literatura de Revisión como Asunto
5.
Rev Med Virol ; 34(2): e2523, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38512106

RESUMEN

COVID-19 is not only associated with substantial acute liver and kidney injuries, but also with an elevated risk of post-acute sequelae involving the kidney and liver system. We aimed to investigate whether COVID-19 exposure increases the long-term risk of kidney and liver disease, and what are the magnitudes of these associations. We searched PubMed, Embase, Web of Science, ClinicalTrials.gov, and the Living Overview of the Evidence COVID-19 Repository for cohort studies estimating the association between COVID-19 and kidney and liver outcomes. Random-effects meta-analyses were performed to combine the results of the included studies. We assessed the certainty of the evidence using the Grading of Recommendations Assessment, Development and Evaluation approach. Fifteen cohort studies with more than 32 million participants were included in the systematic review COVID-19 was associated with a 35% greater risk of kidney diseases (10 more per 1000 persons; low certainty evidence) and 54% greater risk of liver disease (3 more per 1000 persons; low certainty evidence). The absolute increases due to COVID-19 for acute kidney injury, chronic kidney disease, and liver test abnormality were 3, 8, and 3 per 1000 persons, respectively. Subgroup analyses found no differences between different type of kidney and liver diseases. The findings provide further evidence for the association between COVID-19 and incident kidney and liver conditions. The absolute magnitude of the effect of COVID-19 on kidney and liver outcomes was, however, relatively small.


Asunto(s)
COVID-19 , Hepatopatías , SARS-CoV-2 , Humanos , COVID-19/epidemiología , COVID-19/complicaciones , Hepatopatías/epidemiología , Hepatopatías/etiología , Hepatopatías/virología , Enfermedades Renales/epidemiología , Enfermedades Renales/etiología , Enfermedades Renales/virología , Factores de Riesgo , Lesión Renal Aguda/epidemiología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/virología
7.
Mol Ther ; 32(4): 1061-1079, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38382529

RESUMEN

Complement-mediated diseases can be treated using systemic inhibitors. However, complement components are abundant in circulation, affecting systemic inhibitors' exposure and efficacy. Furthermore, because of complement's essential role in immunity, systemic treatments raise infection risk in patients. To address these challenges, we developed antibody fusion proteins combining the alternative-pathway complement inhibitor factor H (fH1-5) with an anti-C3d monoclonal antibody (C3d-mAb-2fH). Because C3d is deposited at sites of complement activity, this molecule localizes to tissue complement while minimizing circulating complement engagement. These fusion proteins bind to deposited complement in diseased human skin sections and localize to activated complement in a primate skin injury model. We further explored the pharmacology of C3d-mAb-2fH proteins in rodent models with robust tissue complement activation. Doses of C3d-mAb-2fH >1 mg/kg achieved >75% tissue complement inhibition in mouse and rat injury models while avoiding circulating complement blockade. Glomerular-specific complement inhibition reduced proteinuria and preserved podocyte foot-process architecture in rat membranous nephropathy, indicating disease-modifying efficacy. These data indicate that targeting local tissue complement results in durable and efficacious complement blockade in skin and kidney while avoiding systemic inhibition, suggesting broad applicability of this approach in treating a range of complement-mediated diseases.


Asunto(s)
Factor H de Complemento , Enfermedades Renales , Humanos , Ratones , Ratas , Animales , Factor H de Complemento/genética , Complemento C3d/metabolismo , Enfermedades Renales/etiología , Anticuerpos , Activación de Complemento
8.
J Allergy Clin Immunol ; 154(1): 237-242.e1, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38555979

RESUMEN

BACKGROUND: Prior studies have reported that renal insufficiency occurs in a small percentage of patients with predominantly antibody deficiency (PAD) and in about 2% of patients with common variable immunodeficiency. OBJECTIVE: The goal of our study was to understand and evaluate the prevalence and type of renal complications in patients with PAD in the United States Immunodeficiency Network (USIDNET) cohort. We hypothesized that there is an association between certain renal complications and severity of immunophenotype in patients with PAD. METHODS: We performed a query of patients with PAD from the USIDNET cohort with renal complications. Patients with documented renal disease such as chronic kidney disease (CKD), nephrolithiasis, nephritis, and renal failure syndrome were included. We compared immunophenotype, flow cytometry findings, and immunoglobulin levels of patients with PAD accompanied by renal complications with those of the total USIDNET cohort of patients with PAD. RESULTS: We determined that 140 of 2071 patients with PAD (6.8%) had renal complications. Of these 140 patients, 50 (35.7%) had CKD, 46 (32.9%) had nephrolithiasis, 18 (12.9 %) had nephritis, and 50 (35.7%) had other renal complications. Compared with the total USIDNET cohort of patients with PAD, patients with CKD had lower absolute lymphocyte counts, CD3+ T-cell counts, CD4+ T-cell counts, CD19+ B-cell counts, CD20+ B-cell counts, and CD27+IgD- B-cell counts (P < .05 for all). Patients with nephritis had lower absolute lymphocyte counts, CD19+ B-cell counts, CD27+ B-cell counts, and IgE levels (P < .05 for all) than patients with PAD without renal disease. CONCLUSIONS: We determined that 6.8% of the USIDNET cohort of patients with PAD had a documented renal complication. Compared with the overall cohort of patients with PAD, those patients with nephritis and CKD had a more severe immunophenotype.


Asunto(s)
Síndromes de Inmunodeficiencia , Humanos , Masculino , Femenino , Estados Unidos/epidemiología , Adulto , Persona de Mediana Edad , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/epidemiología , Síndromes de Inmunodeficiencia/complicaciones , Enfermedades Renales/inmunología , Enfermedades Renales/etiología , Enfermedades Renales/epidemiología , Prevalencia , Adolescente , Inmunofenotipificación , Adulto Joven , Anciano , Estudios de Cohortes , Niño
9.
Am J Physiol Cell Physiol ; 327(4): C994-C1011, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39183639

RESUMEN

The increasing prevalence of obesity-related glomerulopathy (ORG) poses a significant threat to public health. Sodium-glucose cotransporter-2 (SGLT2) inhibitors effectively reduce body weight and total fat mass in individuals with obesity and halt the progression of ORG. However, the underlying mechanisms of their reno-protective effects in ORG remain unclear. We established a high-fat diet-induced ORG model using C57BL/6J mice, which were divided into three groups: normal chow diet (NCD group), high-fat diet (HFD) mice treated with placebo (ORG group), and HFD mice treated with empagliflozin (EMPA group). We conducted 16S ribosomal RNA gene sequencing of feces and analyzed metabolites from kidney, feces, liver, and serum samples. ORG mice showed increased urinary albumin creatinine ratio, cholesterol, triglyceride levels, and glomerular diameter compared with NCD mice (all P < 0.05). EMPA treatment significantly alleviated these parameters (all P < 0.05). Multitissue metabolomics analysis revealed lipid metabolic reprogramming in ORG mice, which was significantly altered by EMPA treatment. MetOrigin analysis showed a close association between EMPA-related lipid metabolic pathways and gut microbiota alterations, characterized by reduced abundances of Firmicutes and Desulfovibrio and increased abundance of Akkermansia (all P < 0.05). The metabolic homeostasis of ORG mice, especially in lipid metabolism, was disrupted and closely associated with gut microbiota alterations, contributing to the progression of ORG. EMPA treatment improved kidney function and morphology by regulating lipid metabolism through the gut-kidney axis, highlighting a novel therapeutic approach for ORG. NEW & NOTEWORTHY Our study uncovered that empagliflozin (EMPA) potentially protects renal function and morphology in obesity-related glomerulopathy (ORG) mice by regulating the gut-kidney axis. EMPA's reno-protective effects in ORG mice are associated with the lipid metabolism, especially in glycerophospholipid metabolism and the pantothenate/CoA synthesis pathways. EMPA's modulation of gut microbiota appears to be pivotal in suppressing glycerol 3-phosphate and CoA synthesis. The insights into gut microbiota-host metabolic interactions offer a novel therapeutic approach for ORG.


Asunto(s)
Compuestos de Bencidrilo , Dieta Alta en Grasa , Microbioma Gastrointestinal , Glucósidos , Riñón , Ratones Endogámicos C57BL , Obesidad , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Glucósidos/farmacología , Glucósidos/uso terapéutico , Compuestos de Bencidrilo/farmacología , Obesidad/metabolismo , Obesidad/tratamiento farmacológico , Ratones , Masculino , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/prevención & control , Enfermedades Renales/etiología , Enfermedades Renales/tratamiento farmacológico , Metabolismo de los Lípidos/efectos de los fármacos , Modelos Animales de Enfermedad
10.
J Lipid Res ; 65(9): 100610, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39094771

RESUMEN

Dyslipidemia may induce chronic kidney disease and trigger both ferroptosis and endoplasmic reticulum (ER) stress, but the instigating factors are incompletely understood. We tested the hypothesis that different models of dyslipidemia engage distinct kidney injury mechanisms. Wild-type (WT) or proprotein-convertase subtilisin/kexin type-9 (PCSK9)-gain-of-function (GOF) Ossabaw pigs were fed with a 6-month normal diet (ND) or high-fat diet (HFD) (n = 5-6 each). Renal function and fat deposition were studied in vivo using CT, and blood and kidney tissue studied ex-vivo for lipid profile, systemic and renal vein FFAs levels, and renal injury mechanisms including lipid peroxidation, ferroptosis, and ER stress. Compared with WT-ND pigs, both HFD and PCSK9-GOF elevated triglyceride levels, which were highest in WT-HFD, whereas total and LDL cholesterol levels rose only in PCSK9-GOF pigs, particularly in PCSK9-GOF/HFD. The HFD groups had worse kidney function than the ND groups. The WT-HFD kidneys retained more FFA than other groups, but all kidneys developed fibrosis. Furthermore, HFD-induced ferroptosis in WT-HFD indicated by increased free iron, lipid peroxidation, and decreased glutathione peroxidase-4 mRNA expression, while PCSK9-GOF induced ER stress with upregulated GRP94 and CHOP protein expression. In vitro, pig kidney epithelial cells treated with palmitic acid and oxidized LDL to mimic HFD and PCSK9-GOF showed similar trends to those observed in vivo. Taken together, HFD-induced hypertriglyceridemia promotes renal FFA retention and ferroptosis, whereas PCSK9-GOF-induced hypercholesterolemia elicits ER stress, both resulting in renal fibrosis. These observations suggest different targets for preventing and treating renal fibrosis in subjects with specific types of dyslipidemia.


Asunto(s)
Dislipidemias , Estrés del Retículo Endoplásmico , Ferroptosis , Fibrosis , Animales , Porcinos , Dislipidemias/metabolismo , Dislipidemias/patología , Riñón/metabolismo , Riñón/patología , Dieta Alta en Grasa/efectos adversos , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/etiología
11.
J Cell Mol Med ; 28(17): e70090, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39261902

RESUMEN

Mitochondrial dysfunction is a pivotal event contributing to the development of ageing-related kidney disorders. Lon protease 1 (LONP1) has been reported to be responsible for ageing-related renal fibrosis; however, the underlying mechanism(s) of LONP1-driven kidney ageing with respect to mitochondrial disturbances remains to be further explored. The level of LONP1 was tested in the kidneys of aged humans and mice. Renal fibrosis and mitochondrial quality control were confirmed in the kidneys of aged mice. Effects of LONP1 silencing or overexpression on renal fibrosis and mitochondrial quality control were explored. In addition, N6-methyladenosine (m6A) modification and methyltransferase like 3 (METTL3) levels, the relationship between LONP1 and METTL3, and the impacts of METTL3 overexpression on mitochondrial functions were confirmed. Furthermore, the expression of insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) and the regulatory effects of IGF2BP2 on LONP1 were confirmed in vitro. LONP1 expression was reduced in the kidneys of aged humans and mice, accompanied by renal fibrosis and mitochondrial dysregulation. Overexpression of LONP1 alleviated renal fibrosis and maintained mitochondrial homeostasis, while silencing of LONP1 had the opposite effect. Impaired METTL3-m6A signalling contributed at least in part to ageing-induced LONP1 modification, reducing subsequent degradation in an IGF2BP2-dependent manner. Moreover, METTL3 overexpression alleviated proximal tubule cell injury, preserved mitochondrial stability, inhibited LONP1 degradation, and protected mitochondrial functions. LONP1 mediates mitochondrial function in kidney ageing and that targeting LONP1 may be a potential therapeutic strategy for improving ageing-related renal fibrosis.


Asunto(s)
Adenosina , Envejecimiento , Fibrosis , Homeostasis , Enfermedades Renales , Riñón , Metiltransferasas , Mitocondrias , Proteínas Mitocondriales , Proteínas de Unión al ARN , Mitocondrias/metabolismo , Animales , Metiltransferasas/metabolismo , Metiltransferasas/genética , Humanos , Envejecimiento/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Riñón/patología , Riñón/metabolismo , Masculino , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/etiología , Enfermedades Renales/genética , Proteasas ATP-Dependientes/metabolismo , Proteasas ATP-Dependientes/genética , Transducción de Señal , Ratones Endogámicos C57BL
12.
Am J Physiol Renal Physiol ; 327(1): F21-F36, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38695077

RESUMEN

According to the Developmental Origins of Health and Disease hypothesis, exposure to certain environmental influences during early life may be a key determinant of fetal development and short- and long-term offspring health. Indeed, adverse conditions encountered during the fetal, perinatal, and early childhood stages can alter normal development and growth, as well as put the offspring at elevated risk of developing long-term health conditions in adulthood, including chronic kidney disease and cardiovascular diseases. Of relevance in understanding the mechanistic basis of these long-term health conditions are previous findings showing low glomerular number in human intrauterine growth restriction and low birth weight-indicators of a suboptimal intrauterine environment. In different animal models, the main suboptimal intrauterine conditions studied relate to maternal dietary manipulations, poor micronutrient intake, prenatal ethanol exposure, maternal diabetes, glucocorticoid and chemical exposure, hypoxia, and placental insufficiency. These studies have demonstrated changes in kidney structure, glomerular endowment, and expression of key genes and signaling pathways controlling endocrine, excretion, and filtration function of the offspring. This review aims to summarize those studies to uncover the effects and mechanisms by which adverse gestational environments impact offspring renal and vascular health in adulthood. This is important for identifying agents and interventions that can prevent and mitigate the long-term consequences of an adverse intrauterine environment on the subsequent generation.NEW & NOTEWORTHY Human data and experimental animal data show that suboptimal environments during fetal development increase the risk of renal and vascular diseases in adult-life. This is related to permanent changes in kidney structure, function, and expression of genes and signaling pathways controlling filtration, excretion, and endocrine function. Uncovering the mechanisms by which offspring renal development and function is impacted is important for identifying ways to mitigate the development of diseases that strain health care services worldwide.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Femenino , Animales , Desarrollo Fetal , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/etiología , Factores de Riesgo
13.
Kidney Int ; 106(2): 214-225, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38797324

RESUMEN

Environmental pollution significantly impacts global disease burden. However, the contribution of environmental pollution to kidney disease is often overlooked in nephrology. This review examines the growing body of research demonstrating the significant impacts of environmental pollutants, with a focus on air pollution as a primary factor, and acknowledges the roles of other pollutants, such as heavy metals, in the development and progression of kidney diseases. Short-term exposure to air pollution is linked with an increased risk of kidney disease-related events, including hospital admissions, and death, predominantly occurring in vulnerable populations. In contrast, long-term exposure, even at low to moderate levels, may lead to progressive pathophysiological changes, such as chronic systemic inflammation and oxidative stress, that contribute to the development of kidney disease. In addition, air pollution may exacerbate traditional kidney disease risk factors such as hypertension and diabetes, thereby accelerating disease progression. The review also explores how climate change may interact with various pollutants, including air pollution, influencing kidney disease indirectly. The examined evidence underscores the urgent need for an interdisciplinary approach to research further into environmental kidney disease. Environmental health policies could play a crucial role in the prevention, intervention, and improvement of kidney health worldwide.


Asunto(s)
Contaminación del Aire , Cambio Climático , Exposición a Riesgos Ambientales , Enfermedades Renales , Humanos , Factores de Riesgo , Exposición a Riesgos Ambientales/efectos adversos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/epidemiología , Enfermedades Renales/etiología , Contaminación del Aire/efectos adversos , Progresión de la Enfermedad , Contaminantes Atmosféricos/efectos adversos , Contaminación Ambiental/efectos adversos
14.
Kidney Int ; 106(5): 840-855, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39181396

RESUMEN

The transcription factor Twist1 plays a vital role in normal development in many tissue systems and continues to be important throughout life. However, inappropriate Twist1 activity has been associated with kidney injury and fibrosis, though the underlying mechanisms involved remain incomplete. Here, we explored the role of Twist1 in regulating fibroblast behaviors and the development kidney fibrosis. Initially Twist1 protein and activity was found to be markedly increased within interstitial myofibroblasts in fibrotic kidneys in both humans and rodents. Treatment of rat kidney interstitial fibroblasts with transforming growth factor-ß1 (a profibrotic factor) also induced Twist1 expression in vitro. Gain- and loss-of-function experiments supported that Twist1 signaling was responsible for transforming growth factor-ß1-induced fibroblast activation and fetal bovine serum-induced fibroblast proliferation. Mechanistically, Twist1 protein promoted kidney fibroblast activation by driving the expression of downstream signaling proteins, Prrx1 and Tnc. Twist1 directly enhanced binding to the promoter of Prrx1 but not TNC, whereas the promoter of TNC was directly bound by Prrx1. Finally, mice with fibroblast-specific deletion of Twist1 exhibited less Prrx1 and TNC protein abundance, interstitial extracellular matrix deposition and kidney inflammation in both the unilateral ureteral obstruction and ischemic-reperfusion injury-induced-kidney fibrotic models. Inhibition of Twist1 signaling with Harmine, a ß-carboline alkaloid, improved extracellular matrix deposition in both injury models. Thus, our results suggest that Twist1 signaling promotes the activation and proliferation of kidney fibroblasts, contributing to the development of interstitial fibrosis, offering a potential therapeutic target for chronic kidney disease.


Asunto(s)
Fibrosis , Proteínas de Homeodominio , Riñón , Proteínas Nucleares , Transducción de Señal , Proteína 1 Relacionada con Twist , Animales , Proteína 1 Relacionada con Twist/metabolismo , Proteína 1 Relacionada con Twist/genética , Humanos , Riñón/patología , Riñón/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Masculino , Ratones , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Miofibroblastos/metabolismo , Miofibroblastos/patología , Proliferación Celular , Factor de Crecimiento Transformador beta1/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/etiología , Ratas , Modelos Animales de Enfermedad , Ratones Noqueados , Fibroblastos/metabolismo , Fibroblastos/patología , Ratones Endogámicos C57BL , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/patología , Obstrucción Ureteral/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/metabolismo , Células Cultivadas
15.
Kidney Int ; 106(1): 98-114, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38521405

RESUMEN

Epigenetic regulations, including DNA methylation, are critical to the development and progression of kidney fibrosis, but the underlying mechanisms remain elusive. Here, we show that fibrosis of the mouse kidney was associated with the induction of DNA methyltransferases and increases in global DNA methylation and was alleviated by the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (5-Aza). Genome-wide analysis demonstrated the hypermethylation of 94 genes in mouse unilateral ureteral obstruction kidneys, which was markedly reduced by 5-Aza. Among these genes, Hoxa5 was hypermethylated at its gene promoter, and this hypermethylation was associated with reduced HOXA5 expression in fibrotic mouse kidneys after ureteral obstruction or unilateral ischemia-reperfusion injury. 5-Aza prevented Hoxa5 hypermethylation, restored HOXA5 expression, and suppressed kidney fibrosis. Downregulation of HOXA5 was verified in human kidney biopsies from patients with chronic kidney disease and correlated with the increased kidney fibrosis and DNA methylation. Kidney fibrosis was aggravated by conditional knockout of Hoxa5 and alleviated by conditional knockin of Hoxa5 in kidney proximal tubules of mice. Mechanistically, we found that HOXA5 repressed Jag1 transcription by directly binding to its gene promoter, resulting in the suppression of JAG1-NOTCH signaling during kidney fibrosis. Thus, our results indicate that loss of HOXA5 via DNA methylation contributes to fibrogenesis in kidney diseases by inducing JAG1 and consequent activation of the NOTCH signaling pathway.


Asunto(s)
Metilación de ADN , Fibrosis , Proteínas de Homeodominio , Proteína Jagged-1 , Regiones Promotoras Genéticas , Receptores Notch , Transducción de Señal , Obstrucción Ureteral , Animales , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Masculino , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/patología , Obstrucción Ureteral/genética , Obstrucción Ureteral/metabolismo , Receptores Notch/metabolismo , Receptores Notch/genética , Riñón/patología , Riñón/metabolismo , Ratones Noqueados , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Epigénesis Genética , Enfermedades Renales/patología , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/etiología , Factores de Transcripción
16.
Mol Med ; 30(1): 174, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39390361

RESUMEN

A new population of peripheral helper T (Tph) cells has been identified and contributed to various autoimmune diseases. Tph cells can secrete interleukin-21 (IL-21), interferon (IFN) and C-X-C motif chemokine ligand 13 (CXCL13) to moderate renal disease. Moreover, Tph cells can congregate in huge numbers and immerse within inflamed tissue. Compared to Tfh cells, Tph cells express high programmed cell death protein 1 (PD-1), major histocompatibility complex II (MHC-II), C-C chemokine receptor 2 (CCR2) and C-C chemokine receptor 5 (CCR5) but often lack expression of the chemokine receptor C-X-C chemokine receptor 5 (CXCR5). They display features distinct from other T cells, which are uniquely poised to promote responses and antibody production of B cells within pathologically inflamed non-lymphoid tissues and a key feature of Tph cells. In this review, we summarize recent findings on the role of Tph cells in chronic kidney disease, acute kidney injury, kidney transplantation and various renal diseases.


Asunto(s)
Enfermedades Renales , Linfocitos T Colaboradores-Inductores , Humanos , Enfermedades Renales/metabolismo , Enfermedades Renales/etiología , Enfermedades Renales/patología , Enfermedades Renales/inmunología , Animales , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Susceptibilidad a Enfermedades , Biomarcadores
17.
Br J Haematol ; 205(4): 1260-1261, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39117989

RESUMEN

The study of Ellsworth et al. (Br J Haematol, 2024) demonstrated the usefulness of oxygen gradient ektacytometry technique to better identify the physiological parameters that could increase the risk of sickling of red blood cells (RBCs) from sickle cell trait (SCT) carriers. Oxygen gradient ektacytometry combined with pH and osmolality modulations could help in identifying SCT carriers at risk for kidney disorders or exercise-related complications. Other factors than the percentages of haemoglobin S are probably involved in the propensity of RBCs from SCT carriers to sickle during deoxygenation. Commentary on: Ellsworth et al. Hypertonicity and/or acidosis induce marked rheological changes under hypoxic conditions in sickle trait red blood cells. Br J Haematol 2024; 205:1565-1569.


Asunto(s)
Ejercicio Físico , Enfermedades Renales , Oxígeno , Rasgo Drepanocítico , Humanos , Rasgo Drepanocítico/genética , Oxígeno/metabolismo , Enfermedades Renales/etiología , Enfermedades Renales/diagnóstico , Eritrocitos/metabolismo , Heterocigoto
18.
Curr Opin Rheumatol ; 36(5): 322-327, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38752864

RESUMEN

PURPOSE OF REVIEW: This review will provide updates in the outcomes in the common rheumatologic diseases with kidney involvement. Covered are also advances in therapeutics for the use of pediatric rheumatologic diseases with kidney involvement, as well as the potential kidney complications from other rheumatologic diseases and their medications. RECENT FINDINGS: Two of the more common rheumatologic diseases with kidney involvement, lupus and vasculitis, continue to show inadequate response to initial therapy of renal disease and practice continues to be driven by results of adult studies. SUMMARY: There is a continued need for pediatric specific studies in rheumatologic diseases with kidney involvement as outcomes continue to be inadequate. Despite recently approved treatments for adults with rheumatic diseases and kidney involvement, therapeutic options in pediatrics remain limited, contributing to the overall morbidity and mortality.


Asunto(s)
Enfermedades Renales , Enfermedades Reumáticas , Humanos , Niño , Enfermedades Reumáticas/complicaciones , Enfermedades Reumáticas/terapia , Enfermedades Renales/etiología , Enfermedades Renales/terapia , Reumatología/métodos , Reumatología/tendencias , Vasculitis/terapia , Vasculitis/etiología , Vasculitis/diagnóstico , Antirreumáticos/uso terapéutico
19.
Biochem Biophys Res Commun ; 715: 149997, 2024 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-38678782

RESUMEN

The immune system is involved in hypertension development with different immune cells reported to have either pro or anti-hypertensive effects. In hypertension, immune cells have been thought to infiltrate blood pressure-regulating organs, resulting in either elevation or reduction of blood pressure. There is controversy over whether macrophages play a detrimental or beneficial role in the development of hypertension, and the few existing studies have yielded conflicting results. This study aimed to determine the effects of angiotensin II (Ang II) salt-induced hypertension on renal immune cells and to determine whether renal macrophages are involved in the induction of hypertension. Hypertension was induced by administration of Ang II and saline for two weeks. The effects of hypertension on kidney immune cells were assessed using flow cytometry. Macrophage infiltration in the kidney was assessed by immunohistochemistry and kidney fibrosis was assessed using trichrome stain and kidney real time-qPCR. Liposome encapsulated clodronate was used to deplete macrophages in C57BL/6J mice and investigate the direct role of macrophages in hypertension induction. Ang II saline mice group developed hypertension, had increased renal macrophages, and had increased expression of Acta2 and Col1a1 and kidney fibrotic areas. Macrophage depletion blunted hypertension development and reduced the expression of Acta2 and Col1a1 in the kidney and kidney fibrotic areas in Ang II saline group. The results of this study demonstrate that macrophages infiltrate the kidneys and increase kidney fibrosis in Ang II salt-induced hypertension, and depletion of macrophages suppresses the development of hypertension and decreases kidney fibrosis. This indicates that macrophages play a direct role in hypertension development. Hence macrophages have a potential to be considered as therapeutic target in hypertension management.


Asunto(s)
Angiotensina II , Modelos Animales de Enfermedad , Fibrosis , Hipertensión , Riñón , Macrófagos , Animales , Ratones , Angiotensina II/farmacología , Presión Sanguínea/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/patología , Hipertensión/metabolismo , Riñón/patología , Riñón/metabolismo , Riñón/efectos de los fármacos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/etiología , Macrófagos/metabolismo , Macrófagos/patología , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Cloruro de Sodio Dietético/efectos adversos
20.
Am J Kidney Dis ; 83(3): 329-339, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37741608

RESUMEN

RATIONALE & OBJECTIVE: Outcomes of kidney transplantation for patients with renal AA amyloidosis are uncertain, with reports of poor survival and high rates of disease recurrence. However, the data are inconclusive and mostly based on studies from the early 2000s and earlier. STUDY DESIGN: Retrospective multicenter cohort study. SETTING & PARTICIPANTS: We searched the French national transplant database to identify all patients with renal AA amyloidosis who underwent kidney transplantation between 2008 and 2018. EXPOSURES: Age, cause of amyloidosis, use of biotherapies, and C-reactive protein levels. OUTCOMES: Outcomes were all-cause mortality and allograft loss. We also reported amyloidosis allograft recurrence, occurrence of acute rejection episodes, as well as infectious, cardiovascular, and neoplastic disease events. ANALYTICAL APPROACH: Kaplan-Meier estimator for mortality and cumulative incidence function method for allograft loss. Factors associated with patient and allograft survival were investigated using a Cox proportional hazards model and a cause-specific hazards model, respectively. RESULTS: 86 patients who received kidney transplants for AA amyloidosis at 26 French centers were included. The median age was 49.4 years (IQR, 39.7-61.1). The main cause of amyloidosis was familial Mediterranean fever (37 cases; 43%). 16 (18.6%) patients received biotherapy after transplantation. Patient survival rates were 94.0% (95% CI, 89.1-99.2) at 1 year and 85.5% (77.8-94.0) at 5 years after transplantation. Cumulative incidences of allograft loss were 10.5% (4.0-17.0) at 1 year and 13.0% (5.8-20.1) at 5 years after transplantation. Histologically proven AA amyloidosis recurrence occurred in 5 transplants (5.8%). An infection requiring hospitalization developed in 55.8% of cases, and there was a 27.9% incidence of acute allograft rejection. Multivariable analysis showed that C-reactive protein concentration at the time of transplantation was associated with patient survival (HR, 1.01; 95% CI, 1.00-1.02; P=0.01) and allograft survival (HR, 1.68; 95% CI, 1.10-2.57; P=0.02). LIMITATIONS: The study lacked a control group, and the effect of biotherapies on transplantation outcomes could not be explored. CONCLUSIONS: This relatively contemporary cohort of patients who received a kidney transplant for AA amyloidosis experienced favorable rates of survival and lower recurrence rates than previously reported. These data support the practice of treating these patients with kidney transplantation for end-stage kidney disease. PLAIN-LANGUAGE SUMMARY: AA amyloidosis is a severe and rare disease. Kidney involvement is frequent and leads to end-stage kidney disease. Because of the involvement of other organs, these patients are often frail, which has raised concerns about their suitability for kidney transplantation. We reviewed all patients with AA amyloidosis nephropathy who underwent kidney transplantation in France in the recent era (2008-2018) and found that the outcomes after kidney transplantation were favorable, with 85.5% of patients still alive 5 years after transplantation, a survival rate that is comparable to the outcomes of patients receiving a transplant for other forms of kidney diseases. Recurrence of amyloidosis in the transplanted kidney was infrequent (5.8%). These data support the practice of kidney transplantation for patients with AA amyloidosis who experience kidney failure.


Asunto(s)
Amiloidosis , Enfermedades Renales , Fallo Renal Crónico , Trasplante de Riñón , Humanos , Persona de Mediana Edad , Trasplante de Riñón/métodos , Estudios de Cohortes , Proteína C-Reactiva , Estudios Retrospectivos , Amiloidosis/cirugía , Amiloidosis/complicaciones , Fallo Renal Crónico/cirugía , Fallo Renal Crónico/complicaciones , Enfermedades Renales/etiología , Estudios Multicéntricos como Asunto , Proteína Amiloide A Sérica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA