Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Kidney Int ; 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38286178

RESUMO

Current classification of chronic kidney disease (CKD) into stages using indirect systemic measures (estimated glomerular filtration rate (eGFR) and albuminuria) is agnostic to the heterogeneity of underlying molecular processes in the kidney thereby limiting precision medicine approaches. To generate a novel CKD categorization that directly reflects within kidney disease drivers we analyzed publicly available transcriptomic data from kidney biopsy tissue. A Self-Organizing Maps unsupervised artificial neural network machine-learning algorithm was used to stratify a total of 369 patients with CKD and 46 living kidney donors as healthy controls. Unbiased stratification of the discovery cohort resulted in identification of four novel molecular categories of disease termed CKD-Blue, CKD-Gold, CKD-Olive, CKD-Plum that were replicated in independent CKD and diabetic kidney disease datasets and can be further tested on any external data at kidneyclass.org. Each molecular category spanned across CKD stages and histopathological diagnoses and represented transcriptional activation of distinct biological pathways. Disease progression rates were highly significantly different between the molecular categories. CKD-Gold displayed rapid progression, with significant eGFR-adjusted Cox regression hazard ratio of 5.6 [1.01-31.3] for kidney failure and hazard ratio of 4.7 [1.3-16.5] for composite of kidney failure or a 40% or more eGFR decline. Urine proteomics revealed distinct patterns between the molecular categories, and a 25-protein signature was identified to distinguish CKD-Gold from other molecular categories. Thus, patient stratification based on kidney tissue omics offers a gateway to non-invasive biomarker-driven categorization and the potential for future clinical implementation, as a key step towards precision medicine in CKD.

2.
J Am Soc Nephrol ; 35(2): 149-165, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38062563

RESUMO

SIGNIFICANCE STATEMENT: Renal fibrosis is a common pathologic process of progressive CKD. We have provided strong evidence that PGI 2 is an important component in the kidney injury/repairing process by reducing fibrosis and protecting renal function from declining. In our study, administration of a PGI 2 analog or selective PTGIR agonist after the acute injury ameliorated renal fibrosis. Our findings provide new insights into the role of PGI 2 in kidney biology and suggest that targeting PGI 2 /PTGIR may be a potential therapeutic strategy for CKD. BACKGROUND: Prostanoids have been demonstrated to be important modulators to maintain tissue homeostasis in response to physiologic or pathophysiologic stress. Prostacyclin (PGI 2 ) is a member of prostanoids. While limited studies have shown that PGI 2 is involved in the tissue injury/repairing process, its role in renal fibrosis and CKD progression requires further investigation. METHODS: Prostacyclin synthase ( Ptgis )-deficient mice, prostaglandin I 2 receptor ( Ptgir )-deficient mice, and an oral PGI 2 analog and selective PTGIR agonist were used to examine the role of PGI 2 in renal fibrosis in mouse models. We also analyzed the single-cell RNA-Seq data to examine the PTGIR -expressing cells in the kidneys of patients with CKD. RESULTS: Increased PTGIS expression has been observed in fibrotic kidneys in both humans and mice. Deletion of the PTGIS gene aggravated renal fibrosis and decline of renal function in murine models. A PGI 2 analog or PTGIR agonist that was administered after the acute injury ameliorated renal fibrosis. PTGIR, the PGI 2 receptor, deficiency blunted the protective effect of the PGI 2 analog. Fibroblasts and myofibroblasts were the major cell types expressing PTGIR in the kidneys of patients with CKD. Deletion of PTGIR in collagen-producing fibroblastic cells aggravated renal fibrosis. The protective effect of PGI 2 was associated with the inhibition of fibroblast activation through PTGIR-mediated signaling. CONCLUSIONS: PGI 2 is an important component in the kidney injury/repairing process by preventing the overactivation of fibroblasts during the repairing process and protecting the kidney from fibrosis and decline of renal function. Our findings suggest that PGI 2 /PTGIR is a potential therapeutic target for CKD.


Assuntos
Epoprostenol , Insuficiência Renal Crônica , Humanos , Animais , Camundongos , Epoprostenol/farmacologia , Epoprostenol/metabolismo , Prostaglandinas I , Rim/metabolismo , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/etiologia , Fibroblastos/metabolismo , Fibrose
3.
J Am Soc Nephrol ; 34(6): 935-936, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37093623
4.
Cell Metab ; 34(7): 1064-1078.e6, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35709763

RESUMO

Diabetic kidney disease (DKD) occurs in ∼40% of patients with diabetes and causes kidney failure, cardiovascular disease, and premature death. We analyzed the response of a murine DKD model to five treatment regimens using single-cell RNA sequencing (scRNA-seq). Our atlas of ∼1 million cells revealed a heterogeneous response of all kidney cell types both to DKD and its treatment. Both monotherapy and combination therapies targeted differing cell types and induced distinct and non-overlapping transcriptional changes. The early effects of sodium-glucose cotransporter-2 inhibitors (SGLT2i) on the S1 segment of the proximal tubule suggest that this drug class induces fasting mimicry and hypoxia responses. Diabetes downregulated the spliceosome regulator serine/arginine-rich splicing factor 7 (Srsf7) in proximal tubule that was specifically rescued by SGLT2i. In vitro proximal tubule knockdown of Srsf7 induced a pro-inflammatory phenotype, implicating alternative splicing as a driver of DKD and suggesting SGLT2i regulation of proximal tubule alternative splicing as a potential mechanism of action for this drug class.


Assuntos
Doenças Cardiovasculares , Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Inibidores do Transportador 2 de Sódio-Glicose , Animais , Doenças Cardiovasculares/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/genética , Camundongos , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Transcriptoma/genética
5.
Prostaglandins Other Lipid Mediat ; 159: 106621, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35131551

RESUMO

Diabetic nephropathy (DN) is a major microvascular complication of diabetes and the leading cause of mortality in diabetic patients. Cyclooxygenase (COX) and COX-derived prostanoids are documented to participate in the pathogenesis of diabetic nephropathy. Herein, we found an increased COX2 expression level in diabetic kidneys of STZ-induced DBA mice. The COX2 inhibitor significantly attenuated albuminuria and histological lesions, accompanied by up-regulation of the renal angiopoietin-1/tie-2 system. This finding is consistent with the presence of an angiogenic signature in endothelial cells during the development of DN. Prostaglandin E2 (PGE2) is the most abundant prostanoid in the kidney, and its receptor EP4 is expressed in the glomerulus, as determined by in situ hybridization. To test the hypothesis that diabetes-associated COX2 overexpression induces renal PGE2 production and endothelial dysfunction by activating glomerular EP4 receptors, the effect of an EP4 antagonist on Akita/DBA mice was investigated. Our results showed that blockade of EP4 receptor significantly reduced albuminuria in diabetic mice. Owing to the established adverse effect of COX2 inhibitors, our study provided new insight into meaningful renal benefits for diabetic nephropathy by targeting the EP4 receptor.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Albuminúria , Animais , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Diabetes Mellitus Experimental/complicações , Dinoprostona , Células Endoteliais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Prostaglandinas , Receptores de Prostaglandina E Subtipo EP4
7.
Diabetes Care ; 43(11): 2760-2767, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32887710

RESUMO

OBJECTIVE: The role of fibrosis in early progressive renal decline in type 2 diabetes is unknown. Circulating WFDC2 (WAP four-disulfide core domain protein 2) and matrix metalloproteinase 7 (MMP-7; Matrilysin) are postulated to be biomarkers of renal fibrosis. This study examined an association of circulating levels of these proteins with early progressive renal decline. RESEARCH DESIGN AND METHODS: Individuals with type 2 diabetes enrolled in the Joslin Kidney Study with an estimated glomerular filtration rate (eGFR) ≥60 mL/min/1.73 m2 were monitored for 6-12 years to ascertain fast early progressive renal decline, defined as eGFR loss ≥5 mL/min/1.73 m2/year. RESULTS: A total of 1,181 individuals were studied: 681 without and 500 with albuminuria. Median eGFR and albumin-to-creatinine ratio (ACR) at baseline were 97 mL/min/1.73 m2 and 24 mg/g, respectively. During follow-up, 152 individuals experienced fast early progressive renal decline: 6.9% in those with normoalbuminuria and 21% with albuminuria. In both subgroups, the risk of renal decline increased with increasing baseline levels of WFDC2 (P < 0.0001) and MMP-7 (P < 0.0001). After adjustment for relevant clinical characteristics and known biomarkers, an increase by one quartile in the fibrosis index (combination of levels of WFDC2 and MMP-7) was associated with higher risk of renal decline (odds ratio 1.63; 95% CI 1.30-2.04). The association was similar and statistically significant among patients with and without albuminuria. CONCLUSIONS: Elevation of circulating profibrotic proteins is associated with the development of early progressive renal decline in type 2 diabetes. This association is independent from albuminuria status and points to the importance of the fibrotic process in the development of early renal decline.


Assuntos
Albuminúria/diagnóstico , Biomarcadores/sangue , Diabetes Mellitus Tipo 2/diagnóstico , Nefropatias Diabéticas/diagnóstico , Metaloproteinase 7 da Matriz/sangue , Proteína 2 do Domínio Central WAP de Quatro Dissulfetos/metabolismo , Adulto , Albuminúria/sangue , Albuminúria/complicações , Biomarcadores/análise , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/patologia , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/patologia , Progressão da Doença , Feminino , Fibrose/sangue , Fibrose/complicações , Fibrose/diagnóstico , Taxa de Filtração Glomerular , Humanos , Rim/patologia , Falência Renal Crônica/sangue , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/etiologia , Falência Renal Crônica/patologia , Estudos Longitudinais , Masculino , Metaloproteinase 7 da Matriz/análise , Metaloproteinase 7 da Matriz/metabolismo , Pessoa de Meia-Idade , New England , Prognóstico , Proteína 2 do Domínio Central WAP de Quatro Dissulfetos/análise
8.
Clin J Am Soc Nephrol ; 15(9): 1342-1351, 2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32193173

RESUMO

As opposed to diseases such as cancer, autoimmune disease, and diabetes, identifying drugs to treat CKD has proven significantly more challenging. Over the past 2 decades, new potential therapeutic targets have been identified as genetically altered proteins involved in rare monogenetic kidney diseases. Other possible target genes have been implicated through common genetic polymorphisms associated with CKD in the general population. Significant challenges remain before translating these genetic insights into clinical therapies for CKD. This paper will discuss how genetic variants may be leveraged to develop drugs and will especially focus on those genes associated with CKD to exemplify the value and challenges in including genetic information in the drug development pipeline.


Assuntos
Desenvolvimento de Medicamentos , Descoberta de Drogas , Genômica , Mutação , Polimorfismo Genético , Fármacos Renais/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Animais , Predisposição Genética para Doença , Humanos , Fenótipo , Fármacos Renais/efeitos adversos , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/genética
9.
J Exp Med ; 216(4): 936-949, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30886059

RESUMO

Diabetic nephropathy is a leading cause of end-stage kidney failure. Reduced angiopoietin-TIE2 receptor tyrosine kinase signaling in the vasculature leads to increased vascular permeability, inflammation, and endothelial cell loss and is associated with the development of diabetic complications. Here, we identified a mechanism to explain how TIE2 signaling is attenuated in diabetic animals. Expression of vascular endothelial protein tyrosine phosphatase VE-PTP (also known as PTPRB), which dephosphorylates TIE2, is robustly up-regulated in the renal microvasculature of diabetic rodents, thereby reducing TIE2 activity. Increased VE-PTP expression was dependent on hypoxia-inducible factor transcriptional activity in vivo. Genetic deletion of VE-PTP restored TIE2 activity independent of ligand availability and protected kidney structure and function in a mouse model of severe diabetic nephropathy. Mechanistically, inhibition of VE-PTP activated endothelial nitric oxide synthase and led to nuclear exclusion of the FOXO1 transcription factor, reducing expression of pro-inflammatory and pro-fibrotic gene targets. In sum, we identify inhibition of VE-PTP as a promising therapeutic target to protect the kidney from diabetic injury.


Assuntos
Nefropatias Diabéticas/metabolismo , Receptor TIE-2/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Animais , Linhagem Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteína Forkhead Box O1/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/metabolismo , RNA Interferente Pequeno/genética
10.
Int J Med Sci ; 16(1): 180-188, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30662341

RESUMO

Aim: Autosomal dominant polycystic kidney disease is one of the most common genetic renal diseases. Cyclooxygenase plays an important role in epithelial cell proliferation and may contribute to the mechanisms underlying cyst formation. The aim of the present study was to evaluate the role of cyclooxygenase inhibition in the cyst progression in polycystic kidney disease. Method: Pkd2WS25/- mice, a murine model which harbors a compound cis-heterozygous mutation of the Pkd2 gene were used. Cyclooxygenase expression was assessed in both human and murine kidney specimens. Pkd2WS25/- mice were treated with Sulindac (a nonselective cyclooxygenase inhibitor) or vehicle for 8 months starting at three weeks age, and then renal cyst burden was assessed by kidney weight and volume. Results: Cyclooxygenase-2 expression was up-regulated compared to control kidneys as shown by RNase protection in human polycystic kidneys and immunoblot in mouse Pkd2WS25/- kidneys. Cyclooxygenase-2 expression was up-regulated in the renal interstitium as well as focal areas of the cystic epithelium (p<0.05). Basal Cyclooxygenase-1 levels were unchanged in both immunohistochemistry and real-time PCR. Administration of Sulindac to Pkd2WS25/- mice and to control mice for 8 months resulted in reduced kidney weights and volume in cystic mice. Renal function and electrolytes were not significantly different between groups. Conclusion: Thus treatment of a murine model of polycystic kidney disease with Sulindac results in decreased kidney cyst burden. These findings provide additional implications for the use of Cyclooxygenase inhibition as treatment to slow the progression of cyst burden in patients with polycystic kidney disease.


Assuntos
Inibidores de Ciclo-Oxigenase/uso terapêutico , Rim Policístico Autossômico Dominante/tratamento farmacológico , Sulindaco/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Cistos/metabolismo , Cistos/fisiopatologia , Dinoprostona/biossíntese , Modelos Animais de Doenças , Progressão da Doença , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Camundongos , Terapia de Alvo Molecular , Mutação , Prostaglandina-E Sintases/biossíntese , Prostaglandina-Endoperóxido Sintases/biossíntese , Prostaglandinas/biossíntese , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
11.
Toxicol Pathol ; 46(8): 991-998, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30392455

RESUMO

The ReninAAV db/db uNx model of diabetic kidney disease (DKD) exhibits hallmarks of advanced human disease, including progressive elevations in albuminuria and serum creatinine, loss of glomerular filtration rate, and pathological changes. Microarray analysis of renal transcriptome changes were more similar to human DKD when compared to db/db eNOS-/- model. The model responds to treatment with arterial pressure lowering (lisinopril) or glycemic control (rosiglitazone) at early stages of disease. We hypothesized the ReninAAV db/db uNx model with advanced disease would have residual disease after treatment with lisinopril, rosiglitazone, or combination of both. To test this, ReninAAV db/db uNx mice with advanced disease were treated with lisinopril, rosiglitazone, or combination of both for 10 weeks. All treatment groups showed significant lowering of urinary albumin to creatinine ratio compared to baseline; however, only combination group exhibited lowering of serum creatinine. Treatment improved renal pathological scores compared to baseline values with residual disease evident in all treatment groups when compared to db/m controls. Gene expression analysis by TaqMan supported pathological changes with increased fibrotic and inflammatory markers. The results further validate this model of DKD in which residual disease is present when treated with agents to lower arterial pressure and glycemic control.


Assuntos
Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Modelos Animais de Doenças , Animais , Humanos , Camundongos , Camundongos Endogâmicos , Transcriptoma
12.
Kidney Int ; 93(5): 1198-1206, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29398132

RESUMO

To identify determinants of early progressive renal decline in type 2 diabetes a range of markers was studied in 1032 patients enrolled into the 2nd Joslin Kidney Study. eGFR slopes estimated from serial measurements of serum creatinine during 5-12 years of follow-up were used to define early renal decline. At enrollment, all patients had normal eGFR, 58% had normoalbuminuria and 42% had albuminuria. Early renal decline developed in 6% and in 18% patients, respectively. As determinants, we examined baseline values of clinical characteristics, circulating markers: TNFR1, KIM-1, and FGF23, and urinary markers: albumin, KIM-1, NGAL, MCP-1, EGF (all normalized to urinary creatinine) and the ratio of EGF to MCP-1. In univariate analysis, all plasma and urinary markers were significantly associated with risk of early renal decline. When analyzed together, systolic blood pressure, TNFR1, KIM-1, the albumin to creatinine ratio, and the EGF/MCP-1 ratio remained significant with the latter having the strongest effect. Integration of these markers into a multi-marker prognostic test resulted in a significant improvement of discriminatory performance of risk prediction of early renal decline, compared with the albumin to creatinine ratio and systolic blood pressure alone. However, the positive predictive value was only 50% in albuminuric patients. Thus, markers in plasma and urine indicate that the early progressive renal decline in Type 2 diabetes has multiple determinants with strong evidence for involvement of tubular damage. However, new, more informative markers are needed to develop a better prognostic test for such decline that can be used in a clinical setting.


Assuntos
Biomarcadores , Diabetes Mellitus Tipo 2/diagnóstico , Nefropatias Diabéticas/etiologia , Adulto , Albuminúria/diagnóstico , Albuminúria/etiologia , Albuminúria/fisiopatologia , Biomarcadores/sangue , Biomarcadores/urina , Pressão Sanguínea , Quimiocina CCL2/urina , Creatinina/urina , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/urina , Nefropatias Diabéticas/diagnóstico , Nefropatias Diabéticas/fisiopatologia , Progressão da Doença , Diagnóstico Precoce , Fator de Crescimento Epidérmico/urina , Feminino , Fator de Crescimento de Fibroblastos 23 , Taxa de Filtração Glomerular , Receptor Celular 1 do Vírus da Hepatite A/sangue , Humanos , Rim/fisiopatologia , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Medição de Risco , Fatores de Risco , Fatores de Tempo
13.
J Am Soc Nephrol ; 29(2): 477-491, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29061652

RESUMO

Progress in research and developing therapeutics to prevent diabetic kidney disease (DKD) is limited by a lack of animal models exhibiting progressive kidney disease. Chronic hypertension, a driving factor of disease progression in human patients, is lacking in most available models of diabetes. We hypothesized that superimposition of hypertension on diabetic mouse models would accelerate DKD. To test this possibility, we induced persistent hypertension in three mouse models of type 1 diabetes and two models of type 2 diabetes by adeno-associated virus delivery of renin (ReninAAV). Compared with LacZAAV-treated counterparts, ReninAAV-treated type 1 diabetic Akita/129 mice exhibited a substantial increase in albumin-to-creatinine ratio (ACR) and serum creatinine level and more severe renal lesions. In type 2 models of diabetes (C57BKLS db/db and BTBR ob/ob mice), compared with LacZAAV, ReninAAV induced significant elevations in ACR and increased the incidence and severity of histopathologic findings, with increased serum creatinine detected only in the ReninAAV-treated db/db mice. The uninephrectomized ReninAAV db/db model was the most progressive model examined and further characterized. In this model, separate treatment of hyperglycemia with rosiglitazone or hypertension with lisinopril partially reduced ACR, consistent with independent contributions of these disorders to renal disease. Microarray analysis and comparison with human DKD showed common pathways affected in human disease and this model. These results identify novel models of progressive DKD that provide researchers with a facile and reliable method to study disease pathogenesis and support the development of therapeutics.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/etiologia , Modelos Animais de Doenças , Hipertensão/complicações , Renina/genética , Animais , Anti-Hipertensivos/uso terapêutico , Creatinina/sangue , Dependovirus , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/patologia , Progressão da Doença , Feminino , Vetores Genéticos , Hipertensão/tratamento farmacológico , Hipertensão/genética , Hipoglicemiantes/uso terapêutico , Janus Quinases/metabolismo , Óperon Lac/genética , Lisinopril/uso terapêutico , Masculino , Camundongos , Nefrectomia , Óxido Nítrico Sintase Tipo III/genética , Rosiglitazona/uso terapêutico , Fatores de Transcrição STAT/metabolismo , Albumina Sérica/metabolismo , Índice de Gravidade de Doença , Transdução de Sinais
14.
Expert Opin Drug Discov ; 13(1): 65-74, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29145743

RESUMO

INTRODUCTION: Diabetic kidney disease (DKD) has emerged as major cause of morbidity and mortality. After progressing to renal failure, over 70% of DKD patients are dead with five years. New treatments to slow this progression are desperately needed. Areas covered: This review highlights the current treatment options for people with DKD with a particular focus on angiotensin pathway blockade and the potential use of sodium glucose linked transporter 2 (SGLT2) inhibitors. These treatments are associated with an initial decrease in glomerular filtration rate (GFR) and albuminuria; there is also attention on renal hyperfiltration as therapeutic target. Both clinical and preclinical testing are facilitated by leveraging albuminuria reduction as a dynamic biomarker of drug effect linked to renal failure. It is critical to ensure that animal models exhibit both albuminuria and progressive loss of renal function so drug effects can be established in both. Expert opinion: New pathways and potential drug targets are emerging from gene expression profiling of human kidney biopsies and genome wide association studies. By harmonizing animal experimentation endpoints with clinical outcomes, focusing on disease pathophysiology and incorporating novel gene expression and biomarker changes, therapeutics can be advanced into clinical testing with greater confidence.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Desenho de Fármacos , Descoberta de Drogas/métodos , Animais , Biomarcadores/metabolismo , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Perfilação da Expressão Gênica , Taxa de Filtração Glomerular , Humanos , Inibidores do Transportador 2 de Sódio-Glicose
15.
Kidney Int ; 92(1): 258-266, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28396115

RESUMO

Design of Phase III trials for diabetic nephropathy currently requires patients at a high risk of progression defined as within three years of a hard end point (end-stage renal disease, 40% loss of estimated glomerular filtration rate, or death). To improve the design of these trials, we used natural history data from the Joslin Kidney Studies of chronic kidney disease in patients with diabetes to develop an improved criterion to identify such patients. This included a training cohort of 279 patients with type 1 diabetes and 134 end points within three years, and a validation cohort of 221 patients with type 2 diabetes and 88 end points. Previous trials selected patients using clinical criteria for baseline urinary albumin-to-creatinine ratio and estimated glomerular filtration rate. Application of these criteria to our cohort data yielded sensitivities (detection of patients at risk) of 70-80% and prognostic values of only 52-63%. We applied classification and regression trees analysis to select from among all clinical characteristics and markers the optimal prognostic criterion that divided patients with type 1 diabetes according to risk. The optimal criterion was a serum tumor necrosis factor receptor 1 level over 4.3 ng/ml alone or 2.9-4.3 ng/ml with an albumin-to-creatinine ratio over 1900 mg/g. Remarkably, this criterion produced similar results in both type 1 and type 2 diabetic patients. Overall, sensitivity and prognostic value were high (72% and 81%, respectively). Thus, application of this criterion to enrollment in future clinical trials could reduce the sample size required to achieve adequate statistical power for detection of treatment benefits.


Assuntos
Ensaios Clínicos Fase III como Assunto/métodos , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/etiologia , Determinação de Ponto Final , Taxa de Filtração Glomerular , Falência Renal Crônica/etiologia , Rim/fisiopatologia , Seleção de Pacientes , Adulto , Albuminúria/etiologia , Albuminúria/fisiopatologia , Biomarcadores/sangue , Biomarcadores/urina , Creatinina/urina , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 2/diagnóstico , Nefropatias Diabéticas/diagnóstico , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/terapia , Progressão da Doença , Feminino , Humanos , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/fisiopatologia , Falência Renal Crônica/terapia , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Reprodutibilidade dos Testes , Medição de Risco , Fatores de Risco , Fatores de Tempo
16.
Am J Physiol Renal Physiol ; 312(6): F951-F962, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28249836

RESUMO

Transforming growth factor-alpha (TGFA) has been shown to play a role in experimental chronic kidney disease associated with nephron reduction, while its role in diabetic kidney disease (DKD) is unknown. We show here that intrarenal TGFA mRNA expression, as well as urine and serum TGFA, are increased in human DKD. We used a TGFA neutralizing antibody to determine the role of TGFA in two models of renal disease, the remnant surgical reduction model and the uninephrectomized (uniNx) db/db DKD model. In addition, the contribution of TGFA to DKD progression was examined using an adeno-associated virus approach to increase circulating TGFA in experimental DKD. In vivo blockade of TGFA attenuated kidney disease progression in both nondiabetic 129S6 nephron reduction and Type 2 diabetic uniNx db/db models, whereas overexpression of TGFA in uniNx db/db model accelerated renal disease. Therapeutic activity of the TGFA antibody was enhanced with renin angiotensin system inhibition with further improvement in renal parameters. These findings suggest a pathologic contribution of TGFA in DKD and support the possibility that therapeutic administration of neutralizing antibodies could provide a novel treatment for the disease.


Assuntos
Nefropatias Diabéticas/metabolismo , Rim/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Idoso , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Pressão Sanguínea , Células Cultivadas , Dependovirus/genética , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Receptores ErbB/metabolismo , Matriz Extracelular/metabolismo , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Taxa de Filtração Glomerular , Humanos , Hipertensão/complicações , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Rim/cirurgia , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Pessoa de Meia-Idade , Nefrectomia , Fosforilação , Sistema Renina-Angiotensina , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador alfa/antagonistas & inibidores , Fator de Crescimento Transformador alfa/deficiência , Fator de Crescimento Transformador alfa/genética
17.
Semin Nephrol ; 36(6): 436-447, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27987541

RESUMO

Chronic kidney disease (CKD) is a lethal and rapidly increasing burden on society. Despite this, there are relatively few therapies in development for the treatment of CKD. Several recent costly phase 3 trials have failed to provide improved renal outcomes, diminishing interest in pharmaceutical investment. Furthermore, poor patient, physician, and payer awareness of CKD as a diagnosis has contributed to slow trial enrollment and successful implementation of these trials. Nevertheless, several therapeutics remain in development for the treatment of CKD, including mineralocorticoid-receptor antagonists, sodium/glucose cotransporter 2 inhibitors, anti-inflammatory drugs, and drugs that mitigate oxidative injury. Success of future CKD therapeutic trials will depend not only on improved understanding of disease pathogenesis, but also on improved trial enrollment rates, through increasing awareness of this disease by the public, policy makers, and the greater medical community.


Assuntos
Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Nefropatias Diabéticas/tratamento farmacológico , Antagonistas dos Receptores de Endotelina/uso terapêutico , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Inibidores do Transportador 2 de Sódio-Glicose , Anti-Inflamatórios/uso terapêutico , Atrasentana , Compostos Benzidrílicos/uso terapêutico , Canagliflozina/uso terapêutico , Descoberta de Drogas , Glucosídeos/uso terapêutico , Glicosaminoglicanos/uso terapêutico , Humanos , Hipoglicemiantes/uso terapêutico , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/uso terapêutico , Estresse Oxidativo , Piridoxamina/análogos & derivados , Piridoxamina/uso terapêutico , Pirrolidinas/uso terapêutico
18.
Nat Rev Drug Discov ; 15(8): 568-88, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27230798

RESUMO

Chronic kidney disease (CKD) represents a leading cause of death in the United States. There is no cure for this disease, with current treatment strategies relying on blood pressure control through blockade of the renin-angiotensin system. Such approaches only delay the development of end-stage kidney disease and can be associated with serious side effects. Recent identification of several novel mechanisms contributing to CKD development - including vascular changes, loss of podocytes and renal epithelial cells, matrix deposition, inflammation and metabolic dysregulation - has revealed new potential therapeutic approaches for CKD. This Review assesses emerging strategies and agents for CKD treatment, highlighting the associated challenges in their clinical development.


Assuntos
Fármacos Renais/farmacologia , Fármacos Renais/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Animais , Fibrose , Humanos , Camundongos , Circulação Renal/efeitos dos fármacos , Insuficiência Renal Crônica/fisiopatologia
19.
J Am Soc Nephrol ; 27(7): 1902-10, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27127187

RESUMO

Innovation in kidney diseases is not commensurate with the effect of these diseases on human health and mortality or innovation in other key therapeutic areas. A primary cause of the dearth in innovation is that kidney diseases disproportionately affect a demographic that is largely disenfranchised, lacking sufficient advocacy, public attention, and funding. A secondary and likely consequent cause is that the existing infrastructure supporting nephrology research pales in comparison with those for other internal medicine specialties, especially cardiology and oncology. Citing such inequities, however, is not enough. Changing the status quo will require a coordinated effort to identify and redress the existing deficits. Specifically, these deficits relate to the need to further develop and improve the following: understanding of the disease mechanisms and pathophysiology, patient engagement and activism, clinical trial infrastructure, and investigational clinical trial designs as well as coordinated efforts among critical stakeholders. This paper identifies potential solutions to these barriers, some of which are already underway through the Kidney Health Initiative. The Kidney Health Initiative is unique and will serve as a current and future platform from which to overcome these barriers to innovation in nephrology.


Assuntos
Pesquisa Biomédica , Nefropatias , Nefrologia , Pesquisa Biomédica/organização & administração , Ensaios Clínicos como Assunto , Humanos , Nefropatias/diagnóstico , Nefropatias/terapia , Terapias em Estudo
20.
J Clin Invest ; 126(2): 695-705, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26690700

RESUMO

Systemic inflammation causes malaise and general feelings of discomfort. This fundamental aspect of the sickness response reduces the quality of life for people suffering from chronic inflammatory diseases and is a nuisance during mild infections like common colds or the flu. To investigate how inflammation is perceived as unpleasant and causes negative affect, we used a behavioral test in which mice avoid an environment that they have learned to associate with inflammation-induced discomfort. Using a combination of cell-type­specific gene deletions, pharmacology, and chemogenetics, we found that systemic inflammation triggered aversion through MyD88-dependent activation of the brain endothelium followed by COX1-mediated cerebral prostaglandin E2 (PGE2) synthesis. Further, we showed that inflammation-induced PGE2 targeted EP1 receptors on striatal dopamine D1 receptor­expressing neurons and that this signaling sequence induced aversion through GABA-mediated inhibition of dopaminergic cells. Finally, we demonstrated that inflammation-induced aversion was not an indirect consequence of fever or anorexia but that it constituted an independent inflammatory symptom triggered by a unique molecular mechanism. Collectively, these findings demonstrate that PGE2-mediated modulation of the dopaminergic motivational circuitry is a key mechanism underlying the negative affect induced by inflammation.


Assuntos
Encefalopatias/metabolismo , Encéfalo/metabolismo , Dinoprostona/metabolismo , Neurônios Dopaminérgicos/metabolismo , Endotélio Vascular/metabolismo , Transmissão Sináptica , Animais , Encéfalo/patologia , Encefalopatias/genética , Encefalopatias/patologia , Linhagem Celular , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Dinoprostona/genética , Neurônios Dopaminérgicos/patologia , Endotélio Vascular/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...