Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Kidney Int Rep ; 9(4): 1020-1030, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38765567

RESUMO

Introduction: The phase 3 DUPLEX trial is evaluating sparsentan, a novel, nonimmunosuppressive, single-molecule dual endothelin angiotensin receptor antagonist, in patients with focal segmental glomerulosclerosis (FSGS). Methods: DUPLEX (NCT03493685) is a global, multicenter, randomized, double-blind, parallel-group, active-controlled study evaluating the efficacy and safety of sparsentan 800 mg once daily versus irbesartan 300 mg once daily in patients aged 8 to 75 years (USA/UK) and 18 to 75 years (ex-USA/UK) weighing ≥20 kg with biopsy-proven FSGS or documented genetic mutation in a podocyte protein associated with FSGS, and urine protein-to-creatinine ratio (UP/C) ≥1.5 g/g. Baseline characteristics blinded to treatment allocation are reported descriptively. Results: The primary analysis population includes 371 patients (336 adult, 35 pediatric [<18 years]) who were randomized and received study drug (median age, 42 years). Patients were White (73.0%), Asian (13.2%), Black/African American (6.7%), or Other race (7.0%); and from North America (38.8%), Europe (36.1%), South America (12.7%), or Asia Pacific (12.4%). Baseline median UP/C was 3.0 g/g; 42.6% in nephrotic-range (UP/C >3.5 g/g [adults]; >2.0 g/g [pediatrics]). Patients were evenly distributed across estimated glomerular filtration rate (eGFR) categories corresponding to chronic kidney disease (CKD) stages 1 to 3b. Thirty-three patients (9.4% of 352 evaluable samples) had pathogenic or likely pathogenic (P/LP) variants of genes essential to podocyte structural integrity and function, 27 (7.7%) had P/LP collagen gene (COL4A3/4/5) variants, and 14 (4.0%) had high-risk APOL1 genotypes. Conclusions: Patient enrollment in DUPLEX, the largest interventional study in FSGS to date, will enable important characterization of the treatment effect of sparsentan in a geographically broad and clinically diverse FSGS population.

2.
Lancet ; 402(10417): 2077-2090, 2023 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-37931634

RESUMO

BACKGROUND: Sparsentan, a novel, non-immunosuppressive, single-molecule, dual endothelin angiotensin receptor antagonist, significantly reduced proteinuria versus irbesartan, an angiotensin II receptor blocker, at 36 weeks (primary endpoint) in patients with immunoglobulin A nephropathy in the phase 3 PROTECT trial's previously reported interim analysis. Here, we report kidney function and outcomes over 110 weeks from the double-blind final analysis. METHODS: PROTECT, a double-blind, randomised, active-controlled, phase 3 study, was done across 134 clinical practice sites in 18 countries throughout the Americas, Asia, and Europe. Patients aged 18 years or older with biopsy-proven primary IgA nephropathy and proteinuria of at least 1·0 g per day despite maximised renin-angiotensin system inhibition for at least 12 weeks were randomly assigned (1:1) to receive sparsentan (target dose 400 mg oral sparsentan once daily) or irbesartan (target dose 300 mg oral irbesartan once daily) based on a permuted-block randomisation method. The primary endpoint was proteinuria change between treatment groups at 36 weeks. Secondary endpoints included rate of change (slope) of the estimated glomerular filtration rate (eGFR), changes in proteinuria, a composite of kidney failure (confirmed 40% eGFR reduction, end-stage kidney disease, or all-cause mortality), and safety and tolerability up to 110 weeks from randomisation. Secondary efficacy outcomes were assessed in the full analysis set and safety was assessed in the safety set, both of which were defined as all patients who were randomly assigned and received at least one dose of randomly assigned study drug. This trial is registered with ClinicalTrials.gov, NCT03762850. FINDINGS: Between Dec 20, 2018, and May 26, 2021, 203 patients were randomly assigned to the sparsentan group and 203 to the irbesartan group. One patient from each group did not receive the study drug and was excluded from the efficacy and safety analyses (282 [70%] of 404 included patients were male and 272 [67%] were White) . Patients in the sparsentan group had a slower rate of eGFR decline than those in the irbesartan group. eGFR chronic 2-year slope (weeks 6-110) was -2·7 mL/min per 1·73 m2 per year versus -3·8 mL/min per 1·73 m2 per year (difference 1·1 mL/min per 1·73 m2 per year, 95% CI 0·1 to 2·1; p=0·037); total 2-year slope (day 1-week 110) was -2·9 mL/min per 1·73 m2 per year versus -3·9 mL/min per 1·73 m2 per year (difference 1·0 mL/min per 1·73 m2 per year, 95% CI -0·03 to 1·94; p=0·058). The significant reduction in proteinuria at 36 weeks with sparsentan was maintained throughout the study period; at 110 weeks, proteinuria, as determined by the change from baseline in urine protein-to-creatinine ratio, was 40% lower in the sparsentan group than in the irbesartan group (-42·8%, 95% CI -49·8 to -35·0, with sparsentan versus -4·4%, -15·8 to 8·7, with irbesartan; geometric least-squares mean ratio 0·60, 95% CI 0·50 to 0·72). The composite kidney failure endpoint was reached by 18 (9%) of 202 patients in the sparsentan group versus 26 (13%) of 202 patients in the irbesartan group (relative risk 0·7, 95% CI 0·4 to 1·2). Treatment-emergent adverse events were well balanced between sparsentan and irbesartan, with no new safety signals. INTERPRETATION: Over 110 weeks, treatment with sparsentan versus maximally titrated irbesartan in patients with IgA nephropathy resulted in significant reductions in proteinuria and preservation of kidney function. FUNDING: Travere Therapeutics.


Assuntos
Glomerulonefrite por IGA , Falência Renal Crônica , Feminino , Humanos , Masculino , Antagonistas de Receptores de Angiotensina/efeitos adversos , Método Duplo-Cego , Glomerulonefrite por IGA/tratamento farmacológico , Irbesartana/efeitos adversos , Proteinúria/tratamento farmacológico , Resultado do Tratamento , Adulto
3.
Sci Rep ; 13(1): 13985, 2023 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-37633958

RESUMO

CKD progression depends on the activation of an intricate set of hemodynamic and inflammatory mechanisms, promoting renal leukocyte infiltration, inflammation and fibrosis, leading to renal function loss. There are currently no specific drugs to detain renal fibrogenesis, which is a common end-point for different nephropathies. Clinical therapy for CKD is mostly based on the management of hypertension and proteinuria, partially achieved with renin-angiotensin-aldosterone system (RAAS) blockers, and the control of inflammation by immunosuppressive drugs. The aim of the present study was to verify if the administration of tamoxifen (TAM), an estrogen receptor modulator, clinically employed in the treatment of breast cancer and predicted to exert antifibrotic effects, would promote additional benefits when associated to a currently used therapeutic scheme for the conservative management of experimental CKD. Wistar rats underwent the NAME model of hypertensive nephrosclerosis, obtained by daily oral administration of a nitric oxide synthesis inhibitor, associated to dietary sodium overload. The therapeutic association of TAM to losartan (LOS), and mofetil mycophenolate (MMF) effectively reduced the severe hypertension, marked albuminuria and glomerular damage exhibited by NAME animals. Moreover, the association also succeeded in limiting renal inflammation in this model, and promoted further reduction of ECM interstitial accumulation and renal fibrosis, compared to the monotherapies. According to our results, the association of TAM to the currently used conservative treatment of CKD added significant antifibrotic effects both in vivo and in vitro, and may represent an alternative to slow the progression of chronic nephropathy.


Assuntos
Hipertensão , Nefroesclerose , Insuficiência Renal Crônica , Ratos , Animais , Ratos Wistar , Nefroesclerose/tratamento farmacológico , Nefroesclerose/etiologia , Tratamento Conservador , Tamoxifeno/farmacologia , Insuficiência Renal Crônica/tratamento farmacológico , Inflamação
4.
Kidney Int Rep ; 8(5): 1043-1056, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37180506

RESUMO

Introduction: Sparsentan is a novel single-molecule dual endothelin angiotensin receptor antagonist with hemodynamic and anti-inflammatory properties and is not an immunosuppressant. The ongoing phase 3 PROTECT trial examines sparsentan in adults with IgA nephropathy (IgAN). Methods: The PROTECT trial (NCT03762850) is a multicenter, international, randomized, double-blind, parallel-group, active-controlled study. The efficacy and safety of sparsentan versus the active control irbesartan is being evaluated in adults with biopsy-proven IgAN and proteinuria ≥1.0 g/d despite maximized treatment with an angiotensin-converting enzyme inhibitor (ACEi) and/or angiotensin receptor blocker (ARB) for at least 12 weeks. Blinded and aggregated baseline characteristics are reported descriptively and compared to contemporary phase 3 trials with patients with IgAN. Results: The primary analysis population includes 404 patients who were randomized and received study drug (median age, 46 years). Enrolled patients were from Europe (53%), Asia Pacific (27%), and North America (20%). Baseline median urinary protein excretion was 1.8 g/d. The range of estimated glomerular filtration rate (eGFR) was broad with the largest proportion of patients (35%) in chronic kidney disease (CKD) stage 3B. Before transitioning to study medication, mean systolic/diastolic blood pressure was 129/82 mm Hg, with the majority of patients (63.4%) receiving the maximum labeled ACEi or ARB dose. Patients in Asian versus non-Asian regions included a higher percentage of females, had lower blood pressures, and included lower proportions of patients with a history of hypertension and baseline antihypertensive treatment. Conclusions: Patient enrollment in PROTECT, with differing racial backgrounds and across CKD stages, will allow for important characterization of the treatment effect of sparsentan in patients with IgAN with proteinuria at high risk of kidney failure.

5.
Lancet ; 401(10388): 1584-1594, 2023 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-37015244

RESUMO

BACKGROUND: Sparsentan is a novel, non-immunosuppressive, single-molecule, dual endothelin and angiotensin receptor antagonist being examined in an ongoing phase 3 trial in adults with IgA nephropathy. We report the prespecified interim analysis of the primary proteinuria efficacy endpoint, and safety. METHODS: PROTECT is an international, randomised, double-blind, active-controlled study, being conducted in 134 clinical practice sites in 18 countries. The study examines sparsentan versus irbesartan in adults (aged ≥18 years) with biopsy-proven IgA nephropathy and proteinuria of 1·0 g/day or higher despite maximised renin-angiotensin system inhibitor treatment for at least 12 weeks. Participants were randomly assigned in a 1:1 ratio to receive sparsentan 400 mg once daily or irbesartan 300 mg once daily, stratified by estimated glomerular filtration rate at screening (30 to <60 mL/min per 1·73 m2 and ≥60 mL/min per 1·73 m2) and urine protein excretion at screening (≤1·75 g/day and >1·75 g/day). The primary efficacy endpoint was change from baseline to week 36 in urine protein-creatinine ratio based on a 24-h urine sample, assessed using mixed model repeated measures. Treatment-emergent adverse events (TEAEs) were safety endpoints. All endpoints were examined in all participants who received at least one dose of randomised treatment. The study is ongoing and is registered with ClinicalTrials.gov, NCT03762850. FINDINGS: Between Dec 20, 2018, and May 26, 2021, 404 participants were randomly assigned to sparsentan (n=202) or irbesartan (n=202) and received treatment. At week 36, the geometric least squares mean percent change from baseline in urine protein-creatinine ratio was statistically significantly greater in the sparsentan group (-49·8%) than the irbesartan group (-15·1%), resulting in a between-group relative reduction of 41% (least squares mean ratio=0·59; 95% CI 0·51-0·69; p<0·0001). TEAEs with sparsentan were similar to irbesartan. There were no cases of severe oedema, heart failure, hepatotoxicity, or oedema-related discontinuations. Bodyweight changes from baseline were not different between the sparsentan and irbesartan groups. INTERPRETATION: Once-daily treatment with sparsentan produced meaningful reduction in proteinuria compared with irbesartan in adults with IgA nephropathy. Safety of sparsentan was similar to irbesartan. Future analyses after completion of the 2-year double-blind period will show whether these beneficial effects translate into a long-term nephroprotective potential of sparsentan. FUNDING: Travere Therapeutics.


Assuntos
Glomerulonefrite por IGA , Adulto , Humanos , Adolescente , Irbesartana/uso terapêutico , Glomerulonefrite por IGA/tratamento farmacológico , Creatinina/urina , Proteinúria/tratamento farmacológico , Método Duplo-Cego , Resultado do Tratamento
6.
Int J Mol Sci ; 23(7)2022 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-35409202

RESUMO

Diabetes mellitus (DM) burden encompasses diabetic kidney disease (DKD), the leading cause of end-stage renal disease worldwide. Despite compelling evidence indicating that pharmacological intervention curtails DKD progression, the search for non-pharmacological strategies can identify novel targets for drug development against metabolic diseases. One of those emergent strategies comprises the modulation of the intestinal microbiota through fecal transplant from healthy donors. This study sought to investigate the benefits of fecal microbiota transplant (FMT) on functional and morphological parameters in a preclinical model of type 2 DM, obesity, and DKD using BTBRob/ob mice. These animals develop hyperglycemia and albuminuria in a time-dependent manner, mimicking DKD in humans. Our main findings unveiled that FMT prevented body weight gain, reduced albuminuria and tumor necrosis factor-α (TNF-α) levels within the ileum and ascending colon, and potentially ameliorated insulin resistance in BTBRob/ob mice. Intestinal structural integrity was maintained. Notably, FMT was associated with the abundance of the succinate-consuming Odoribacteraceae bacteria family throughout the intestine. Collectively, our data pointed out the safety and efficacy of FMT in a preclinical model of type 2 DM, obesity, and DKD. These findings provide a basis for translational research on intestinal microbiota modulation and testing its therapeutic potential combined with current treatment for DM.


Assuntos
Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Albuminúria/complicações , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Nefropatias Diabéticas/metabolismo , Transplante de Microbiota Fecal/efeitos adversos , Camundongos , Camundongos Endogâmicos , Obesidade/complicações , Obesidade/metabolismo , Obesidade/terapia
7.
J Pharm Pharmacol ; 73(8): 1092-1100, 2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-33950246

RESUMO

OBJECTIVES: The aim of this study was to test whether lipid core nanoparticles loaded with paclitaxel (LDE-PTX) protect rat aortic allograft from immunological damage. METHODS: Fisher and Lewis rats were used differing in minor histocompatibility loci. Sixteen Lewis rats were allocated to four-animal groups: SYNG (syngeneic), Lewis rats receiving aorta grafts from Lewis rats; ALLO (allogeneic), Lewis rats receiving aortas from Fisher rats; ALLO+LDE (allogeneic transplant treated with LDE), Lewis rats receiving aortas from Fisher rats, treated with LDE (weekly injection for 3 weeks); ALLO+LDE-PTX (allogeneic transplant treated with LDE-PTX), Lewis rats receiving aortas from Fisher rats treated with LDE-PTX (4 mg/kg weekly for 3 weeks). Treatments began on transplantation day. RESULTS: Thirty days post-transplantation, SYNG showed intact aortas. ALLO and ALLO+LDE presented intense neointimal formation. In ALLO+LDE-PTX, treatment inhibited neointimal formation; narrowing of aortic lumen was prevented in ALLO and ALLO+LDE. LDE-PTX strongly inhibited proliferation and intimal invasion by smooth muscle cells, diminished 4-fold presence of apoptotic/dead cells in the intima, reduced the invasion of aorta by macrophages and T-cells and gene expression of pro-inflammatory tumour necrosis factor-alpha (TNFα), interferon gamma (IFNγ) and interleukin-1 beta (IL-1ß). CONCLUSIONS: LDE-PTX was effective in preventing the vasculopathy associated with rejection and may offer a potent therapeutic tool for post-transplantation.


Assuntos
Aloenxertos , Aorta/transplante , Sistemas de Liberação de Fármacos por Nanopartículas/farmacologia , Neointima , Paclitaxel/farmacologia , Aloenxertos/metabolismo , Aloenxertos/patologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Proteínas Reguladoras de Apoptose/análise , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Interferon gama/análise , Interleucina-1beta/análise , Neointima/metabolismo , Neointima/patologia , Neointima/prevenção & controle , Ratos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo , Enxerto Vascular/métodos
8.
Sci Rep ; 11(1): 5925, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33723330

RESUMO

Sepsis is the leading cause of acute kidney injury (AKI) and lung injury worldwide. Despite therapeutic advances, sepsis continues to be associated with high mortality. Because Brazilian green propolis (GP) has promising anti-inflammatory, antioxidant, and immunomodulatory properties, we hypothesized that it would protect kidneys and lungs in rats induced to sepsis by cecal ligation and puncture (CLP). Male Wistar rats were divided into groups-control (sham-operated); CLP (CLP only); and CLP + GP (CLP and treatment with GP at 6 h thereafter)-all receiving volume expansion and antibiotic therapy at 6 h after the procedures. By 24 h after the procedures, treatment with GP improved survival, attenuated sepsis-induced AKI, and restored renal tubular function. Whole-blood levels of reduced glutathione were higher in the CLP + GP group. Sepsis upregulated the Toll-like receptor 4/nuclear factor-kappa B axis in lung and renal tissues, as well as increasing inflammatory cytokine levels and macrophage infiltration; all of those effects were attenuated by GP. Treatment with GP decreased the numbers of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling-positive cells in renal and lung tissue, as well as protecting the morphology of the renal mitochondria. Our data open the prospect for clinical trials of the use of GP in sepsis.


Assuntos
Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/prevenção & controle , Anti-Infecciosos/farmacologia , Própole/química , Sepse/complicações , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Anti-Infecciosos/química , Apoptose , Biomarcadores , Quimiotaxia de Leucócito/imunologia , Cromatografia Líquida de Alta Pressão , Citocinas/metabolismo , Modelos Animais de Doenças , Testes de Função Renal , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Ratos , Transdução de Sinais
9.
J. bras. nefrol ; 42(3): 370-374, July-Sept. 2020. graf
Artigo em Inglês, Português | LILACS | ID: biblio-1134841

RESUMO

Abstract Encapsulating Peritoneal Sclerosis (EPS) is a severe and rare condition frequently associated with peritoneal dialysis, characterized by bowel obstruction, with lethal consequences in 20% of the patients. The disease presents as a mass of fibrous tissue encapsulating visceral organs that may potentially compromise digestive tract function. This report describes the case of a patient under peritoneal dialysis (PD) due to chronic kidney disease secondary to focal segmental glomerulosclerosis diagnosed with EPS. The patient had undergone two living-donor kidney transplant procedures. Surgical techniques and clinical measures employed to unravel bowel obstruction are described, which have been shown to ameliorate EPS secondary complications. Parenteral nutrition has significantly contributed to afford adequate nutrition, improving tissue healing as well as serum protein levels, vitamins and electrolytes. Therapy with tamoxifen and sodium thiosulfate effectively delayed the development of EPS.


Resumo A peritonite esclerosante encapsulante (PEE) é uma condição rara e grave, frequentemente associada à diálise peritoneal, caracterizada por obstrução intestinal, que pode ter uma evolução fatal em 20% dos pacientes. Apresenta-se como uma massa de tecido fibroso, recobrindo os tecidos viscerais, e pode comprometer o funcionamento fisiológico de todo o aparelho digestivo. O presente estudo relata um caso de PEE decorrente de diálise peritoneal (DP) devido à insuficiência renal por glomeruloesclerose focal e segmentar. No caso relatado, a paciente foi submetida a DP e passou por dois transplantes renais intervivos. São descritas as técnicas cirúrgicas e as medidas clínicas adotadas, que se revelaram úteis na resolução da obstrução intestinal, com melhora dos efeitos secundários da PEE. A dieta parenteral mostrou ser um importante fator para a manutenção do aporte nutricional, auxiliando na cicatrização e no nível sérico de proteínas, vitaminas e eletrólitos. A terapia com tamoxifeno e a administração de hipossulfito de sódio foram eficientes para retardar o avanço da PEE.


Assuntos
Humanos , Feminino , Pré-Escolar , Adulto , Peritonite , Transplante de Rim , Diálise Peritoneal , Fibrose Peritoneal , Imunossupressores
10.
Am J Physiol Regul Integr Comp Physiol ; 318(1): R135-R147, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31596111

RESUMO

Sepsis induces organ dysfunction due to overexpression of the inflammatory host response, resulting in cardiopulmonary and autonomic dysfunction, thus increasing the associated morbidity and mortality. Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) express genes and secrete factors with anti-inflammatory properties, neurological and immunological protection, as well as improve survival in experimental sepsis. The cholinergic anti-inflammatory pathway (CAP) is mediated by α7-nicotinic acetylcholine receptors (α7nAChRs), which play an important role in the control of systemic inflammation. We hypothesized that WJ-MSCs attenuate sepsis-induced organ injury in the presence of an activated CAP pathway. To confirm our hypothesis, we evaluated the effects of WJ-MSCs as a treatment for cardiopulmonary injury and on neuroimmunomodulation. Male Wistar rats were randomly divided into four groups: control (sham-operated); cecal ligation and puncture (CLP) alone; CLP+WJ-MSCs (1 × 106 cells, at 6 h post-CLP); and CLP+methyllycaconitine (MLA)+WJ-MSCs (5 mg/kg body wt, at 5.5 h post-CLP, and 1 × 106 cells, at 6 h post-CLP, respectively). All experiments, including the assessment of echocardiographic parameters and heart rate variability, were performed 24 h after CLP. WJ-MSC treatment attenuated diastolic dysfunction and restored baroreflex sensitivity. WJ-MSCs also increased cardiac sympathetic and cardiovagal activity. WJ-MSCs reduced leukocyte infiltration and proinflammatory cytokines, effects that were abolished by administration of a selective α7nAChR antagonist (MLA). In addition, WJ-MSC treatment also diminished apoptosis in the lungs and spleen. In cardiac and splenic tissue, WJ-MSCs downregulated α7nAChR expression, as well as reduced the phospho-STAT3-to-total STAT3 ratio in the spleen. WJ-MSCs appear to protect against sepsis-induced organ injury by reducing systemic inflammation, at least in part, via a mechanism that is dependent on an activated CAP.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Neuroimunomodulação , Sepse/terapia , Geleia de Wharton/citologia , Animais , Citocinas , Humanos , Masculino , Miocárdio/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Baço/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
11.
Sci Rep ; 9(1): 19604, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31862892

RESUMO

Podocytes are specialized cells with a limited capacity for cell division that do not regenerate in response to injury and loss. Insults that compromise the integrity of podocytes promote proteinuria and progressive renal disease. The aim of this study was to evaluate the potential renoprotective and regenerative effects of mesenchymal stromal cells (mSC) in a severe form of the podocyte injury model induced by intraperitoneal administration of puromycin, aggravated by unilateral nephrectomy. Bone derived mSC were isolated and characterized according to flow cytometry analyses and to their capacity to differentiate into mesenchymal lineages. Wistar rats were divided into three groups: Control, PAN, and PAN+ mSC, consisting of PAN rats treated with 2 × 105 mSC. PAN rats developed heavy proteinuria, hypertension, glomerulosclerosis and significant effacement of the foot process. After 60 days, PAN rats treated with mSC presented a significant amelioration of all these abnormalities. In addition, mSC treatment recovered WT1 expression, improved nephrin, podocin, synaptopodin, podocalyxin, and VEGF expression, and downregulated proinflammatory Th1 cytokines in the kidney with a shift towards regulatory Th2 cytokines. In conclusion, mSC administration induced protection of podocytes in this experimental PAN model, providing new perspectives for the treatment of renal diseases associated with podocyte damage.


Assuntos
Nefropatias/terapia , Células-Tronco Mesenquimais/citologia , Podócitos/citologia , Animais , Diferenciação Celular , Divisão Celular , Regulação para Baixo , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/urina , Hipertensão , Inflamação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Nefropatias/induzido quimicamente , Masculino , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Nefrectomia , Podócitos/efeitos dos fármacos , Proteinúria/urina , Puromicina Aminonucleosídeo , Ratos , Ratos Wistar , Regeneração , Sialoglicoproteínas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Cell Transplant ; 27(5): 739-753, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29701108

RESUMO

Acute kidney injury (AKI), characterized by a sharp drop in glomerular filtration, continues to be a significant health burden because it is associated with high initial mortality, morbidity, and substantial health-care costs. There is a strong connection between AKI and mechanisms of senescence activation. After ischemic or nephrotoxic insults, a wide range of pathophysiological events occur. Renal tubular cell injury is characterized by cell membrane damage, cytoskeleton disruption, and DNA degradation, leading to tubular cell death by necrosis and apoptosis. The senescence mechanism involves interstitial fibrosis, tubular atrophy, and capillary rarefaction, all of which impede the morphological and functional recovery of the kidneys, suggesting a strong link between AKI and the progression of chronic kidney disease. During abnormal kidney repair, tubular epithelial cells can assume a senescence-like phenotype. Cellular senescence can occur as a result of cell cycle arrest due to increased expression of cyclin kinase inhibitors (mainly p21), downregulation of Klotho expression, and telomere shortening. In AKI, cellular senescence is aggravated by other factors including oxidative stress and autophagy. Given this scenario, the main question is whether AKI can be repaired and how to avoid the senescence process. Stem cells might constitute a new therapeutic approach. Mesenchymal stem cells (MSCs) can ameliorate kidney injury through angiogenesis, immunomodulation, and fibrosis pathway blockade, as well as through antiapoptotic and promitotic processes. Young umbilical cord-derived MSCs are better at increasing Klotho levels, and thus protecting tissues from senescence, than are adipose-derived MSCs. Umbilical cord-derived MSCs improve glomerular filtration and tubular function to a greater degree than do those obtained from adult tissue. Although senescence-related proteins and microRNA are upregulated in AKI, they can be downregulated by treatment with umbilical cord-derived MSCs. In summary, stem cells derived from young tissues, such as umbilical cord-derived MSCs, could slow the post-AKI senescence process.


Assuntos
Injúria Renal Aguda/terapia , Envelhecimento/patologia , Rim/patologia , Injúria Renal Aguda/patologia , Animais , Pontos de Checagem do Ciclo Celular , Progressão da Doença , Glucuronidase/genética , Humanos , Proteínas Klotho
13.
PLoS One ; 12(9): e0184302, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28873458

RESUMO

BACKGROUND: Progressive fibrous thickening of the peritoneal membrane is a complication of long-term peritoneal dialysis (PD). TGF-ß/Smad pathway activation, inflammation, and neoangiogenesis play important roles in peritoneal membrane (PM) changes induced by PD. Recently, histone deacetilase inhibitors (HDACi) have shown anti-fibrotic and anti-inflammatory effects in different experimental models. These drugs prevent deacetylation of histones causing a loosen chromatin, which in turn induce the expression of some anti-fibrotic genes. In addition, acetylation may increase the activity of proteins involved in tissue fibrosis, such as Smad7. Here, we explored the effect of valproic acid (VPA), an HDACi, on the development of peritoneal fibrosis (PF) in rats. METHODS: PF was induced by daily intraperitoneal injections of 0.1% chlorhexidine gluconate (CG) for 15 consecutive days. Male Wistar rats (250-300 g) were divided into 3 groups: CONTROL, control rats receiving only vehicle; PF, peritoneal fibrosis induced in rats; PF+VPA, rats with PF treated with VPA (300 mg/kg/day by gavage). PF was assessed by Masson's trichrome staining. Inflammation and fibrosis-associated factors were assessed by immunohistochemistry, immunofluorescence, multiplex analysis, and qPCR. RESULTS: Treatment with VPA significantly reduced PM thickness and the expression of myofibroblasts, besides preventing loss of ultrafiltration capacity of the PM. The upregulation of profibrotic factors (TGF-ß, fibronectin, and Smad3) in the PF group was significantly ameliorated by VPA. VPA modulated the TGF/Smad pathway, inhibiting phosphorylated Smad3 expression and inducing an increased Smad7 expression in the FP+VPA group. The neoangiogenesis and the expression of proinflammatory cytokines (TNF-α, IL-1ß, MCP-1) observed in the PF group was significantly reduced by VPA. CONCLUSIONS: Our results indicate that VPA suppressed experimental PF through modulation of the TGF-ß/Smad pathway. Interestingly, VPA treatment induced a higher expression of antifibrotic factors, such as Smad7. These results suggest that VPA may represent a potential strategy for treating long term PD complications.


Assuntos
Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/tratamento farmacológico , Ácido Valproico/uso terapêutico , Animais , Transporte Biológico/efeitos dos fármacos , Biomarcadores/metabolismo , Peso Corporal/efeitos dos fármacos , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Capilares/metabolismo , Contagem de Células , Citocinas/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/patologia , Mediadores da Inflamação , Masculino , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Neovascularização Fisiológica , Fibrose Peritoneal/genética , Fibrose Peritoneal/patologia , Peritônio/efeitos dos fármacos , Peritônio/metabolismo , Peritônio/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Resultado do Tratamento , Ácido Valproico/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Biomed Res Int ; 2016: 5985903, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27195290

RESUMO

Ischemia-reperfusion injury (IRI) is a major contributor to acute kidney injury (AKI). At present, there are no effective therapies to prevent AKI. The aim of this study was to analyse whether valproic acid (VPA), a histone deacetylase inhibitor with anti-inflammatory properties, prevents renal IRI. Male Wistar rats were divided into three groups: SHAM rats were subjected to a SHAM surgery, IRI rats underwent bilateral renal ischemia for 45 min, and IRI + VPA rats were treated with VPA at 300 mg/kg twice daily 2 days before bilateral IRI. Animals were euthanized at 48 hours after IRI. VPA attenuated renal dysfunction after ischemia, which was characterized by a decrease in BUN (mg/dL), serum creatinine (mg/dL), and FENa (%) in the IRI + VPA group (39 ± 11, 0.5 ± 0.05, and 0.5 ± 0.06, resp.) compared with the IRI group (145 ± 35, 2.7 ± 0.05, and 4.9 ± 1, resp.; p < 0.001). Additionally, significantly lower acute tubular necrosis grade and number of apoptotic cells were found in the IRI + VPA group compared to the IRI group (p < 0.001). Furthermore, VPA treatment reduced inflammatory cellular infiltration and expression of proinflammatory cytokines. These data suggest that VPA prevents the renal dysfunction and inflammation that is associated with renal IRI.


Assuntos
Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/fisiopatologia , Nefrite/prevenção & controle , Nefrite/fisiopatologia , Traumatismo por Reperfusão/fisiopatologia , Ácido Valproico/administração & dosagem , Injúria Renal Aguda/etiologia , Animais , Anti-Inflamatórios/administração & dosagem , Masculino , Nefrite/etiologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/tratamento farmacológico , Resultado do Tratamento
15.
Oxid Med Cell Longev ; 2016: 2189582, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26881017

RESUMO

Diabetes mellitus leads to increased Advanced Glycation End Products (AGE) production, which has been associated with secondary diabetic complications. Type 1 diabetic patients undergoing pancreas-kidney transplantation (SPKT) can restore normoglycemia and renal function, eventually decreasing AGE accumulation. We aimed to prospectively study AGE evolution after SPKT. Circulating AGE were assessed in 20 patients, at time 0 (T0), 3 months (T3), 6 months (T6), and 12 months (T12) after successful SPKT. Global AGE and carboxymethyllysine (CML) were analyzed, as well as advanced oxidation protein products (AOPP). Skin biopsies were obtained at T0 and T12. Immunohistochemistry with anti-AGE antibody evaluated skin AGE deposition. AGE mean values were 16.8 ± 6.4 µg/mL at T0; 17.1 ± 3.8 µg/mL at T3; 17.5 ± 5.6 µg/mL at T6; and 16.0 ± 5.2 µg/mL at T12. CML mean values were 0.94 ± 0.36 ng/mL at T0; 1.11 ± 0.48 ng/mL at T3; 0.99 ± 0.42 ng/mL at T6; and 0.78 ± 0.38 ng/mL at T12. AOPP mean values were 130.1 ± 76.8 µMol/L at T0; 137.3 ± 110.6 µMol/L at T3; 116.4 ± 51.2 µMol/L at T6; and 106.4 ± 57.9 µMol/L at T12. CML variation was significant (P = 0.022); AOPP variation was nearly significant (P = 0.076). Skin biopsies evolved mostly from a cytoplasmic diffuse to a peripheral interkeratinocytic immunoreaction pattern; in 7 cases, a reduction in AGE immunoreaction intensity was evident at T12. In conclusion, glycoxidation markers decrease, plasmatic and on tissues, may start early after SPKT. Studies with prolonged follow-up may confirm these data.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Transplante de Rim/efeitos adversos , Transplante de Pâncreas/efeitos adversos , Pele/metabolismo , Adulto , Biópsia , Estudos de Casos e Controles , Diabetes Mellitus Tipo 1/complicações , Feminino , Humanos , Imuno-Histoquímica , Terapia de Imunossupressão , Lisina/análogos & derivados , Lisina/química , Masculino , Pessoa de Meia-Idade , Oxigênio/química , Estudos Prospectivos , Proteínas/química , Pele/patologia , Fatores de Tempo
16.
Nephrol Dial Transplant ; 28(5): 1140-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23345625

RESUMO

BACKGROUND: Persistent systemic inflammation has been widely recognized in patients with chronic kidney disease (CKD), and is associated with increased risk of morbidity and mortality. Intervention therapies aiming for the blockade of inflammatory cytokines are considered attractive approaches for CKD patients with signs of chronic inflammation. In this context, thalidomide, due to its potent anti-inflammatory and immunomodulatory properties, may represent an alternative strategy of treatment. In the present study, we developed an experimental model of CKD with uraemia in mice, induced by a diet rich in adenine, which causes progressive renal dysfunction, resembling the human uraemic features. Inflammatory parameters were analysed in this model of CKD and the potential beneficial effects of thalidomide as an anti-inflammatory drug was also investigated. METHODS: C57/BL-6 mice were fed with an adenine-containing diet during a period of 6 weeks. Thirty mice were divided into three groups: Control group (animals receiving normal diet), ADE group (mice receiving adenine-containing diet) and ADE + TLD group (CKD mice receiving thalidomide, 30 mg/kg/day, by gavage). Besides biochemical and histopathological changes, local and systemic inflammatory parameters were also analysed, including expression of cytokines interleukin (IL)-1ß, tumour necrosis factor-α, IL-6, IL-4 and IL-10 in kidney samples by real-time RT-PCR and quantification of serum levels of cytokines. Finally, the electrophoretic mobility shift assay (EMSA) for NF-κB was also examined. RESULTS: Adenine-fed mice developed advanced CKD characterized by a marked increase in serum urea, creatinine, phosphorus and intact parathyroid hormone (iPTH) levels. In addition, histological changes of tubulointerstitial injury, characterized by deposition of crystals in the kidney, accompanied by tubular dilatation, degeneration of proximal tubular epithelium with loss of the brush border, inflammatory cellular infiltration, foreign-body granuloma formation and interstitial fibrosis were also evident. By immunohistochemistry, Mac-2- and α-SMA-positive cells were identified in the tubulointerstitial compartment. Treatment with thalidomide significantly reduced serum urea, creatinine, phosphorus and iPTH levels and protected against tubulointerstitial injury. Local and systemic inflammation in the mice model of adenine-induced CKD was confirmed by the findings of significantly high expression of cytokine mRNA levels and NF-κB activation in the kidney tissue as well as marked increased serum levels of inflammatory cytokines. Thalidomide treatment significantly reduced gene expression of these cytokines and the activation of the NF-κB in the renal tissue and the circulating levels of cytokines. CONCLUSIONS: Dietary adenine caused advanced CKD with uraemia in mice providing a useful experimental model to study molecular and morphological changes associated with this disease. The negative impact of inflammation in this CKD model was overcome by the marked anti-inflammatory effects of thalidomide, promoting renal protection.


Assuntos
Adenina/toxicidade , Modelos Animais de Doenças , Imunossupressores/farmacologia , Inflamação/prevenção & controle , Insuficiência Renal Crônica/complicações , Talidomida/farmacologia , Uremia/complicações , Animais , Western Blotting , Citocinas/genética , Citocinas/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Técnicas Imunoenzimáticas , Inflamação/etiologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Insuficiência Renal Crônica/induzido quimicamente , Insuficiência Renal Crônica/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Uremia/induzido quimicamente , Uremia/patologia
17.
J Am Soc Nephrol ; 23(1): 37-48, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22052053

RESUMO

Tamoxifen, a selective estrogen receptor modulator, has antifibrotic properties; however, whether it can attenuate renal fibrosis is unknown. In this study, we tested the effects of tamoxifen in a model of hypertensive nephrosclerosis (chronic inhibition of nitric oxide synthesis with L-NAME). After 30 days, treated rats had significantly lower levels of albuminuria as well as lower histologic scores for glomerulosclerosis and interstitial fibrosis than untreated controls. Tamoxifen was renoprotective despite having no effect on the sustained, severe hypertension induced by L-NAME. Tamoxifen prevented the accumulation of extracellular matrix by decreasing the expression of collagen I, collagen III, and fibronectin mRNA and protein. These renoprotective effects associated with inhibition of TGF-ß1 and plasminogen activator inhibitor-1, and with a significant reduction in α-smooth muscle actin-positive cells in the renal interstitium. Furthermore, tamoxifen abrogated IL-1ß- and angiotensin-II-induced proliferation of fibroblasts from both kidney explants and from the NRK-49F cell line. Tamoxifen also inhibited the expression of extracellular matrix components and the production and release of TGF-ß1 into the supernatant of these cells. In summary, tamoxifen exhibits antifibrotic effects in the L-NAME model of hypertensive nephrosclerosis, likely through the inhibition of TGF-ß1, suggesting that it may have therapeutic use in CKD treatment.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Fibroblastos/efeitos dos fármacos , Nefropatias/tratamento farmacológico , Tamoxifeno/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Matriz Extracelular/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Rim/metabolismo , Rim/patologia , Nefropatias/patologia , Masculino , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ratos , Ratos Wistar , Tamoxifeno/farmacologia , Fator de Crescimento Transformador beta1/metabolismo
18.
Rev Assoc Med Bras (1992) ; 57(1): 100-6, 2011.
Artigo em Português | MEDLINE | ID: mdl-21390466

RESUMO

This review presents current considerations for renal transplantation in HIV patients. In the last decade, with the advent of highly active antiretroviral therapy (HAART), life expectancy of patients infected with human immunodeficiency virus (HIV) has significantly improved, showing a marked decrease in the rates of morbidity and mortality in this population. In this setting, the number of HIV-positive patients with end stage chronic kidney disease requiring dialysis is progressively growing. Kidney transplantation, previously considered as absolute contraindication for HIV-infected patients is currently, in the HAART era, considered a possible treatment alternative. Concerns for the effects of immunosuppressive drugs in these patients and the possible effects on progression of HIV disease, in addition to the risk of opportunistic infections and cancer development are widely discussed. Clinical experience in the HAART era shows that use of immunosuppressive drugs does not adversely affect HIV-seropositive patients. Furthermore, several transplant centers have reported improved patient and graft outcomes for kidney transplant recipients infected with HIV. In summary, results obtained so far are encouraging, supporting that renal transplantation, following specific selection criteria, can be considered an alternative of renal replacement therapy in HIV-infected patients.


Assuntos
Infecções por HIV/complicações , Falência Renal Crônica/cirurgia , Transplante de Rim/imunologia , Antirretrovirais/uso terapêutico , Terapia Antirretroviral de Alta Atividade , Infecções por HIV/tratamento farmacológico , Humanos , Terapia de Imunossupressão , Imunossupressores/administração & dosagem , Seleção de Pacientes
19.
Rev. Assoc. Med. Bras. (1992) ; 57(1): 100-106, jan.-fev. 2011. tab
Artigo em Português | LILACS | ID: lil-576158

RESUMO

Este artigo de revisão tem como objetivo apresentar as principais considerações sobre o transplante renal em pacientes HIV positivos. Na última década, com o advento da terapia antirretroviral de alta atividade (Highly Active Antiretroviral Therapy - HAART), a evolução dos pacientes infectados pelo vírus da imunodeficiência humana (Human Immunodeficiency Virus - HIV) mudou significativamente, com uma acentuada diminuição das taxas de morbimortalidade nesta população. Neste contexto, o número de pacientes HIV positivos com doença renal crônica com necessidade de terapia dialítica vem aumentando progressivamente. Diante desta nova realidade, o transplante renal, antes considerado uma contraindicação absoluta para tais pacientes, passou a ser considerado uma alternativa de terapia substitutiva da função renal. Questões sobre o uso de imunossupressores neste grupo de pacientes e sua possível ação aumentando a replicação do HIV, além do risco de infecções oportunistas e de desenvolvimento de neoplasias, são amplamente discutidas. Porém, a experiência clínica nessa área mostra que a utilização dessas drogas para pacientes soropositivos parece ser segura, inclusive com relatos de ação antirretroviral de algumas das drogas imunossupressoras. Apesar disso, ainda hoje existem poucos relatos de transplantes nesta população. Em resumo, os dados da literatura sugerem que o transplante renal, seguindo critérios de seleção dos pacientes, parece ser uma alternativa segura como terapia de substituição renal em pacientes HIV positivos.


This review presents current considerations for renal transplantation in HIV patients. In the last decade, with the advent of highly active antiretroviral therapy (HAART), life expectancy of patients infected with human immunodeficiency virus (HIV) has significantly improved, showing a marked decrease in the rates of morbidity and mortality in this population. In this setting, the number of HIV-positive patients with end stage chronic kidney disease requiring dialysis is progressively growing. Kidney transplantation, previously considered as absolute contraindication for HIV-infected patients is currently, in the HAART era, considered a possible treatment alternative. Concerns for the effects of immunosuppressive drugs in these patients and the possible effects on progression of HIV disease, in addition to the risk of opportunistic infections and cancer development are widely discussed. Clinical experience in the HAART era shows that use of immunosuppressive drugs does not adversely affect HIV-seropositive patients. Furthermore, several transplant centers have reported improved patient and graft outcomes for kidney transplant recipients infected with HIV. In summary, results obtained so far are encouraging, supporting that renal transplantation, following specific selection criteria, can be considered an alternative of renal replacement therapy in HIV-infected patients.


Assuntos
Humanos , Infecções por HIV/complicações , Falência Renal Crônica/cirurgia , Transplante de Rim/imunologia , Terapia Antirretroviral de Alta Atividade , Antirretrovirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , Terapia de Imunossupressão , Imunossupressores/administração & dosagem , Seleção de Pacientes
20.
Kidney Int Suppl (2011) ; 1(3): 77-82, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25028628

RESUMO

Amniotic fluid (AF) contains a variety of cell types derived from fetal tissues that can easily grow in culture. These cells can be obtained during amniocentesis for prenatal screening of fetal genetic diseases, usually performed during the second trimester of pregnancy. Of particular interest, some expanded sub-populations derived from AF cells are capable of extensive self-renewal and maintain prolonged undifferentiated proliferation, which are defining properties of stem cells. These human AF stem cells (hAFSCs) exhibit multilineage potential and can differentiate into the three germ layers. They have high proliferation rates and express mesenchymal and embryonic markers, but do not induce tumor formation. In this study, hAFSCs derived from amniocentesis performed at 16-20 weeks of pregnancy were isolated, grown in culture, and characterized by flow cytometry and by their potential ability to differentiate into osteogenic, adipogenic, and chondrogenic lineages. After 4-7 passages, 5 × 105 hAFSCs were inoculated under the kidney capsule of Wistar rats that were subjected to an experimental model of chronic renal disease, the 5/6 nephrectomy model (Nx). After 30 days, Nx rats treated with hAFSCs displayed significant reductions in blood pressure, proteinuria, macrophages, and α-smooth muscle actin expression compared with Nx animals. These preliminary results suggest that hAFSCs isolated and expanded from AF obtained by routine amniocentesis can promote renoprotection in the Nx model. Considering that the AF cells not used for fetal karyotyping are usually discarded, and that their use does not raise ethical issues, they may represent an alternative source of stem cells for cell therapy and regenerative medicine.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA