Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Intervalo de año de publicación
1.
Diabetes Care ; 46(8): 1501-1506, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37267479

RESUMEN

OBJECTIVE: Tirzepatide reduces HbA1c and body weight, and creatinine-based estimated glomerular filtration rate (eGFR) decline. Unlike creatine-derived eGFR (eGFR-creatinine), cystatin C-derived eGFR (eGFR-cystatin C) is unaffected by muscle mass changes. We assessed effects of tirzepatide on eGFR-creatinine and eGFR-cystatin C. RESEARCH DESIGN AND METHODS: Our primary outcome was eGFR change from baseline at 52 weeks with pooled tirzepatide (5, 10, and 15 mg) and titrated insulin glargine in adults with type 2 diabetes and high cardiovascular risk (SURPASS-4). RESULTS: Least squares mean (SE) eGFR-creatinine (mL/min/1.73 m2) changes from baseline with tirzepatide and insulin glargine were -2.5 (0.38) and -3.9 (0.38) (between-group difference, 1.4 [95% CI 0.3-2.4]) and -3.5 (0.37) and -5.3 (0.37) (between-group difference, 1.8 [95% CI 0.8-2.8]) for eGFR-cystatin C. Baseline, 1-year, and 1-year change from baseline values significantly correlated between eGFR-cystatin C and eGFR-creatinine. Measures of eGFR changes did not correlate with body weight changes. CONCLUSIONS: Tirzepatide slows the eGFR decline rate, supporting a kidney-protective effect.


Asunto(s)
Diabetes Mellitus Tipo 2 , Adulto , Humanos , Insulina Glargina/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Cistatina C/farmacología , Creatinina , Tasa de Filtración Glomerular/fisiología , Riñón , Peso Corporal
2.
Oncogene ; 42(3): 209-223, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36402931

RESUMEN

Mast cells (MCs) are abundantly distributed in the human intestinal mucosa and submucosa. However, their roles and mechanisms in the development of colorectal cancer (CRC) are still unclear. In the present research, we found that the infiltration density of MCs in CRC tissues was positively correlated with improved patients' prognoses. Moreover, MCs suppressed the growth and induced the apoptosis of CRC cells in vitro and in vivo but had no effect on normal colonic epithelial cells. The present study revealed that MCs specifically induced endoplasmic reticulum stress (ERS) and activated the unfolded protein response (UPR) in CRC cells but not in normal cells, which led to the suppression of CRC development in vivo. Furthermore, we found that the secreted Cystatin C protein was the key factor for the MC-induced ERS in CRC cells. This work is of significance for uncovering the antitumor function of MCs in CRC progression and identifying the potential of CRC to respond to MC-targeted immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/patología , Mastocitos/metabolismo , Mastocitos/patología , Cistatina C/metabolismo , Cistatina C/farmacología , Estrés del Retículo Endoplásmico , Respuesta de Proteína Desplegada , Proteínas/metabolismo , Apoptosis
3.
PeerJ ; 10: e14232, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36312752

RESUMEN

Background: Periodontal disease is considered one of the most prevalent chronic infectious diseases, often leading to the disruption of tooth-supporting tissues, including alveolar bone, causing tooth mobility and loss. Porphyromonas gingivalis is considered the major etiological agent of this disease, having a plethora of virulence factors, including, lipopolysaccharides (LPS), hemolysins, and proteinases. Antimicrobial peptides are one of the main components of the innate immune response that inhibit the growth of P. gingivalis. The aim of this study was to analyze the antimicrobial activity of cystatin C and to assess the effect on the inflammatory and anti-inflammatory cytokines, the production of reactive oxygen species, and in the release of nitric oxide by human gingival fibroblasts incubated with P. gingivalis in the presence and absence of cystatin C. Methods: P. gingivalis ATCC 33277 was exposed to cystatin C for 24h and co-cultured with human gingival fibroblasts (HGFs) ATCC CRL-2014. The effect of cystatin on growth of P. gingivalis and HGFs was evaluated. Pro-inflammatory (TNFα, IL-1ß) and anti-inflammatory (IL-10) cytokines were determined by ELISA in the supernatants of HGFs incubated with P. gingivalis exposed to cystatin C. Additionally, nitrites and reactive oxygen species (ROS) production were evaluated. Results: Cystatin Cinhibited the growth of P. gingivalis without affecting HGFs. Incubation of HGFs with P. gingivalis led to a significant increase of TNF-α and IL-1ß. In contrast, HGFs incubated with P. gingivalis exposed to cystatin C showed a decreased production of both cytokines, whereas IL-10 was enhanced. Incubation of HGFs with P. gingivalis led to an increase of nitric oxide (NO) and ROS production, which was reduced in the presence of the peptide. Conclusions: Cystatin C inhibits the growth of P. gingivalis and decreases the inflammatory cytokines, ROS, and NO production during infection of HGFs with P. gingivalis. Knowledge on the antimicrobial and immunomodulatory properties of cystatin C could aid in the design of new therapeutic approaches to facilitate the elimination of this bacterium to improve the treatment of periodontal disease.


Asunto(s)
Antiinfecciosos , Enfermedades Periodontales , Humanos , Porphyromonas gingivalis , Interleucina-10/farmacología , Especies Reactivas de Oxígeno/farmacología , Cistatina C/farmacología , Óxido Nítrico/farmacología , Citocinas/farmacología , Antiinflamatorios/farmacología , Antiinfecciosos/farmacología , Fibroblastos
4.
Cardiovasc Drugs Ther ; 36(5): 829-840, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-34232433

RESUMEN

PURPOSE: Ticagrelor and dapagliflozin can suppress the activation of the NOD-like receptor 3 (NLRP3)-inflammasome and activate AMP-activated protein kinase (AMPK). The anti-inflammatory effects of dapagliflozin has been shown to depend on AMPK activation. Dapagliflozin and ticagrelor have been shown to have additive effects on the progression of diabetic cardiomyopathy in BTBR ob/ob mice with type-2 diabetes. We assessed whether dapagliflozin and ticagrelor have additive effects on the activation of the NLRP3-inflammasome and the progression of diabetic nephropathy in mice with type-2 diabetes. METHODS: Eight-week-old BTBR received either no-drug, dapagliflozin (1.5 mg/kg/d), ticagrelor (100 mg/kg/d), or their combination for 12 weeks. Blood was assessed weekly for glucose and urine for glucose and albumin. After 12 weeks, blood creatinine, cystatin C, inflammasome activation, and insulin were assessed by ELISA. Renal cortex samples were assessed by hematoxylin and eosin and periodic acid-Schiff staining. RT-PCR and immunoblotting were used to evaluate fibrosis and the activation of Akt, AMPK and the inflammasome. RESULTS: Both ticagrelor and dapagliflozin reduced serum creatinine and cystatin C levels and urinary albumin. Both drugs attenuated the increase in glomerular area and mesangial matrix index. Both drugs decreased collagen-1 and collagen-3 expression and the activation of the NLRP3-inflammasome. Both drugs increased P-AMPK levels, but only dapagliflozin increased P-Akt levels. Overall, the protective effects of dapagliflozin and ticagrelor were additive. CONCLUSIONS: Dapagliflozin and ticagrelor attenuated the progression of diabetic nephropathy in BTBR ob/ob mice with additive effects of the combination. This was associated with AMPK activation and reduced activation of the NLRP3 inflammasome, whereas only dapagliflozin increased Akt activation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Insulinas , Proteínas Quinasas Activadas por AMP/metabolismo , Albúminas/metabolismo , Albúminas/farmacología , Albúminas/uso terapéutico , Animales , Antiinflamatorios/farmacología , Compuestos de Bencidrilo , Creatinina/metabolismo , Creatinina/farmacología , Creatinina/uso terapéutico , Cistatina C/metabolismo , Cistatina C/farmacología , Cistatina C/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/prevención & control , Eosina Amarillenta-(YS)/metabolismo , Eosina Amarillenta-(YS)/farmacología , Eosina Amarillenta-(YS)/uso terapéutico , Glucosa/metabolismo , Glucósidos , Hematoxilina/metabolismo , Hematoxilina/farmacología , Hematoxilina/uso terapéutico , Inflamasomas/metabolismo , Insulinas/metabolismo , Insulinas/farmacología , Insulinas/uso terapéutico , Riñón , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácido Peryódico/metabolismo , Ácido Peryódico/farmacología , Ácido Peryódico/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ticagrelor/farmacología , Ticagrelor/uso terapéutico
5.
FEBS Open Bio ; 10(10): 2166-2181, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32810913

RESUMEN

Cysteine proteases are implicated in proteolysis events favoring cancer cell growth, spread, and death by apoptosis. Herein, we have studied whether the net growth and survival of the leukemic cell lines Jurkat, U937, and HL-60 are affected by external addition of five proteins acting as natural cysteine protease inhibitors. None of the cystatins examined (A, C, D, and E/M) or chagasin showed consistent effects on Fas-induced apoptosis when evaluated at 1 µm. In contrast, when the intrinsic apoptosis pathway was activated by hydrogen peroxide, addition of cystatin D augmented caspase-3-like activity within all three cell lines. Flow cytometric analysis of U937 cells also showed increased numbers of annexin V-positive cells when hydrogen peroxide was used to initiate apoptosis and cells were cultured in the presence of cystatin D or C. Moreover, stimulation of hydrogen peroxide-induced apoptotic U937 cells with either cystatin C or D resulted in a dose-dependent decrease in the number of cells. Cell viability was also decreased when U937 cells were cultured in the presence of cystatin C or D (1-9 µm) only, demonstrating that these cystatins can reduce cell proliferation by themselves in addition to enhancing apoptosis induced by oxidative stress. These effects on U937 cells were paralleled by internalization of cystatins C and D, indicating these effects are caused by downregulation of intracellular proteolysis. External addition of cystatins C and D to HL-60 and Jurkat cells demonstrated similar degrees of cystatin D uptake and decreased viability as for U937 cells, indicating that these effects are general for leukemic cells.


Asunto(s)
Cistatina C/farmacología , Cistatinas/farmacología , Leucemia/metabolismo , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cistatina C/metabolismo , Cistatinas/metabolismo , Cistatinas/fisiología , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Leucemia/genética , Proteolisis , Transducción de Señal/efectos de los fármacos
6.
Eur Rev Med Pharmacol Sci ; 24(11): 6360-6370, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32572933

RESUMEN

OBJECTIVE: At present, the incidence of acute myocardial infarction is increasing year by year, and it has become one of the diseases with the highest mortality rate in humans. Myocardial ischemia-reperfusion injury (MIRI) is a major problem in the treatment of myocardial infarction, but clinically there is no effective way to treat MIRI. This study used Cystatin C (Cys C) to treat cardiomyocytes and rats to investigate the effect of Cys C on MIRI. MATERIALS AND METHODS: We used H2O2 to induce rat cardiomyocytes (H9c2 cells) injury and stimulated the cells with Cys C. Cell counting kit 8 (CCK8) assay was used to determine the optimal concentration of H2O2 and Cys C to stimulate H9c2 cells. We determined the effects of Cys C on oxidative stress and apoptosis levels in H9c2 cells by measuring the activity of dehydrogenase (LDH), superoxide dismutase (SOD) and malondialdehyde (MDA), and the expression of apoptosis-related molecules (caspase3/8/9, Bax and Bcl-2). Changes in the activity of the NF-κB signaling pathway in H9c2 cells were also detected. In addition, we made rat MIRI models by ligating the coronary arteries and used Cys C to treat rats to verify the effect of Cys C on MIRI. RESULTS: According to the results of the CCK8 assay, 1000 µM of H2O2 and 15 µM of Cys C were used to stimulate H9c2 cells. Cys C alleviated H2O2-induced H9c2 cell injury, manifested as a decrease in LDH and MDA activity and an increase in SOD activity. Cys C also reduced the apoptosis level in H9c2 cells. The activity of NF-κB signaling pathway in injured H9c2 cells was increased, and stimulation of Cys C could inhibit the NF-κB signaling pathway in H9c2 cells. The application of Cys C in MIRI rats also verified its therapeutic effect on MIRI. CONCLUSIONS: Cys C reduced the oxidative stress and apoptosis levels of cardiomyocytes by inhibiting the activity of NF-κB signaling pathway in cardiomyocytes, thereby reducing cardiomyocyte injury and treating MIRI.


Asunto(s)
Cistatina C/farmacología , Peróxido de Hidrógeno/antagonistas & inhibidores , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Cistatina C/administración & dosificación , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/farmacología , Inyecciones Subcutáneas , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
7.
Cell Death Dis ; 9(5): 506, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29724997

RESUMEN

While wound healing is completed, the epithelium functions to normalize the interstitial context by eliminating fibroblasts excited during matrix reconstruction. If not, tissues undergo pathologic fibrosis. Pulmonary fibrosis is a fatal and hardly curable disorder. We here tried to identify epithelium-derived cytokines capable of ameliorating pulmonary fibrosis. Human lung fibroblasts were inactivated in epithelial cell-conditioned media. Cystatin C (CST3) and growth differentiation factor 15 (GDF15) were found to be enriched in the conditioned media and to inhibit the growth and activation of lung fibroblasts by inactivating the TGF-Smad pathway. In mouse and human lungs with interstitial fibrosis, CST3 and GDF15 expressions were markedly reduced, and the restoration of these cytokines alleviated the fibrotic changes in mouse lungs. These results suggest that CST3 and GDF15 are bona fide regulators to prevent excessive proliferation and activation of fibroblasts in injured lungs. These cytokines could be potential therapeutics for ameliorating interstitial lung fibrosis.


Asunto(s)
Cistatina C/genética , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Fibrosis Pulmonar Idiopática/genética , Enfermedades Pulmonares Intersticiales/genética , Células A549 , Animales , Bleomicina/administración & dosificación , Proliferación Celular , Cistatina C/metabolismo , Cistatina C/farmacología , Citocinas/genética , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/farmacología , Células HCT116 , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Pulmón/patología , Enfermedades Pulmonares Intersticiales/metabolismo , Enfermedades Pulmonares Intersticiales/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Análisis de Supervivencia
9.
Biochem Biophys Res Commun ; 500(2): 288-295, 2018 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-29653105

RESUMEN

The final strategies to care patients with end-stage renal fibrosis rely on dialysis and kidney transplantation. Because such treatments are invasive and cause health problems eventually, it is necessary to develop new therapeutic strategies for delaying the disease progress. We here searched for cytokines showing an anti-fibrotic activity in cell-based experiments. Cystatin C (CST3) and Growth differentiation factor 15 (GDF15) were identified to have anti-fibrotic activities in a cytokine array screening. In primary fibroblasts isolated from the mouse kidneys subjected to ureteral obstruction-induced fibrosis, each cytokine induced apoptotic cell death and reduced collagen production. These anti-fibrotic effects were further augmented by co-administration of both cytokines. Mechanistically, CST3 and GDF15 were found to block the TGF-ß receptor and the N-Myc signaling pathways, respectively. In mice with unilateral ureter obstruction, each cytokine and the combination of two cytokines effectively reduced the fibrotic burden in the subjected kidneys. Therefore, we propose that CST3 and GDF15 could be potential candidates for biopharmaceutics to ameliorate renal fibrosis.


Asunto(s)
Cistatina C/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Factor 15 de Diferenciación de Crecimiento/farmacología , Riñón/patología , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno/biosíntesis , Fibroblastos/efectos de los fármacos , Fibrosis , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
10.
Anim Sci J ; 89(7): 1027-1032, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29602234

RESUMEN

A milk protein fraction possessing alkaline isoelectric points (milk basic protein [MBP]) improves bone metabolism in vivo, and it inhibits bone resorption by osteoclasts and promotes mouse osteoblastic MC3T3-E1 cells proliferation in vitro. Cystatin C (CysC) is a component of MBP and shows bone resorption inhibitory activity. Therefore, it is likely that MBP with higher CysC content improves bone metabolism more effectively. In this study, we prepared MBP with low and high contents of CysC and compared its effects on bone metabolism with standard MBP in vitro. Our results showed that the CysC content in MBP was positively related to not only bone resorption inhibitory activity but also MC3T3-E1 cells proliferative activity. Furthermore, purified CysC stimulated MC3T3-E1 cells proliferation. These results indicate that CysC contributes to promotion of MC3T3-E1 cells proliferation, and MBP with higher CysC content shows enhanced bone resorption inhibitory activity and MC3T3-E1 cells proliferative activity. CysC is considered an important factor in the effect on bone metabolism of MBP.


Asunto(s)
Resorción Ósea/prevención & control , Huesos/metabolismo , Proliferación Celular/efectos de los fármacos , Cistatina C/farmacología , Proteínas de la Leche/química , Osteoblastos/fisiología , Osteoclastos/efectos de los fármacos , Animales , Células Cultivadas , Cistatina C/aislamiento & purificación , Cistatina C/fisiología , Relación Dosis-Respuesta a Droga , Ratones , Estimulación Química
11.
Mol Cell Neurosci ; 89: 1-8, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29577984

RESUMEN

In Alzheimer's disease (AD) tau protein hyperphosphorylation causes neurofibrillary tangle formation, microtubule instability and neurodegeneration. Determining the mechanism of tau hyperphosphorylation will provide a better understanding of AD pathology. Cystatin C (CysC) is a risk factor for late-onset AD and its level is upregulated in the brains of AD patients. The role of CysC is AD pathogenesis is not known. In this study, we found that CysC level is upregulated in 3xTg-AD mouse brain. We demonstrate that CysC does not affect cellular Aß production. However, when overexpressed in neuron (NGF-differentiated PC12 cells), CysC inhibits turnover of GSK3ß, promotes GSK3ß-catalyzed tau phosphorylation at Ser396/404 and causes microtubule instability. Our data provide a novel insight into the role of CysC in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Cistatina C/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Microtúbulos/metabolismo , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas tau/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Microtúbulos/efectos de los fármacos , Neuronas/efectos de los fármacos , Células PC12 , Fosforilación , Proteolisis , Ratas
12.
Hemodial Int ; 22(1): 56-65, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28078828

RESUMEN

INTRODUCTION: There is no consensus on the specific indications for weaning critically ill patients with acute kidney injury (AKI) off renal replacement therapy (RRT). This study aimed to explore the prognostic value of several biomarkers measured upon discontinuation of RRT for their value in predicting 60-day survival and renal recovery in an effort to add knowledge to the decision-making process regarding RRT withdrawal. METHODS: We prospectively enrolled 102 patients with AKI who required RRT from the intensive care unit. Serum osteopontin (sOPN), serum interleukin 6 (sIL-6), serum cystatin C (sCysC), sIL-18, serum neutrophil gelatinase-associated lipocalin and urinary IL-18 and urinary neutrophil gelatinase-associated lipocalin were measured upon discontinuation of RRT. Patients were followed up at 60 days for survival and renal recovery. FINDINGS: Patients who survived showed lower levels of all serum and urinary biomarkers. Serum OPN (OR 1.029, 95% CI 1.013-1.047, P = 0.001), diabetes (OR 23.157, 95% CI 4.507-118.981, P < 0.001) and APACHE II score (OR 1.308, 95% CI 1.121-1.527, P = 0.001) were independent predictors of 60-day mortality. Patients whose sOPN values fell within the highest and middle tertiles showed 5.25- and 2.31-fold increased risks of mortality, respectively, compared with that of patients in the lowest tertile. The addition of sOPN to the clinical model resulted in significant net reclassification improvement of 0.453 (P = 0.026) and an integrated discriminative index of 0.155 (P = 0.032). Lower levels of sOPN and sIL-6 were associated with greater odds of 60-day survival (AUC 0.812 and 0.741). The AUC value for predicting survival reached its highest level when all biomarkers were combined with urine output (UO) and urinary and serum creatinine upon discontinuation of RRT (0.882). Lower sCysC performed as well as higher UO in predicting 60-day renal recovery with the greatest AUC of 0.743. DISCUSSION: Upon discontinuation of RRT, serum and urinary biomarkers, particularly sOPN, may predict 60-day survival and renal recovery in critically ill patients with AKI. The serum levels of OPN, IL-6 and CysC may be useful when considering withdrawal of RRT on the basis of conventional indicators.


Asunto(s)
Lesión Renal Aguda/terapia , Biomarcadores/orina , Cistatina C/uso terapéutico , Diálisis Renal/métodos , Terapia de Reemplazo Renal/métodos , Lesión Renal Aguda/mortalidad , Enfermedad Crítica , Cistatina C/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
13.
J Neurochem ; 145(1): 80-89, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29282717

RESUMEN

Cystatin C (CysC) is a major protein component of Bunina bodies, which are a pathological hallmark observed in the remaining motor neurons of patients with amyotrophic lateral sclerosis (ALS). Dominant mutations in the SOD1 gene, encoding Cu/Zn superoxide dismutase (SOD1), are causative for a subset of inherited ALS cases. Our previous study showed that CysC exerts a neuroprotective effect against mutant SOD1-mediated toxicity in vitro; however, in vivo evidence of the beneficial effects mediated by CysC remains obscure. Here we examined the therapeutic potential of recombinant human CysC in vivo using a mouse model of ALS in which the ALS-linked mutated SOD1 gene is expressed (SOD1G93A mice). Intracerebroventricular administration of CysC during the early symptomatic SOD1G93A mice extended their survival times. Administered CysC was predominantly distributed in ventral horn neurons including motor neurons, and induced autophagy through AMP-activated kinase activation to reduce the amount of insoluble mutant SOD1 species. Moreover, PGC-1α, a disease modifier of ALS, was restored by CysC through AMP-activated kinase activation. Finally, the administration of CysC also promoted aggregation of CysC in motor neurons, which is similar to Bunina bodies. Taken together, our findings suggest that CysC represents a promising therapeutic candidate for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Cistatina C/farmacología , Neuronas Motoras/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Autofagia/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación , Proteínas Recombinantes/farmacología , Superóxido Dismutasa-1/genética
14.
Cell Death Dis ; 8(6): e2854, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28569795

RESUMEN

Cystatin C (CYS C, Cst3) is an endogenous cysteine protease inhibitor that plays neuroprotective roles in neurodegenerative diseases. We aimed to explore the association of CYS C with Parkinson's disease (PD) models and investigate its involvement in the role of neurovascular units (NVUs) in PD neuro-pathogenesis. We used A53T α-synuclein (SNCA) transgenic mice and 6-hydroxydopamine-lesioned DAergic PC12 cells as experimental PD models to investigate the mechanisms behind this association. The injections of CYS C were administered to the right substantia nigra (SN) of A53T SNCA transgenic mice to measure the effects of CYS C in transgenic A53T SNCA mice. To explore the angiogenesis in vivo and in vitro, we used the chick embryo chorioallantoic membrane (CAM) assay and tube formation (TF) assay. We found that CYS C has a neuroprotective effect in this in vivo PD model. We observed increased VEGF, NURR1 and autophagy markers LC3B and decreased SNCA and apoptosis marker cleaved CASP3 in different brain regions of CYS C-treated A53T SNCA transgenic mice. In vitro, we observed that CYS C-induced VEGF, a secreted protein, attenuated 6-OHDA-lesioned DAergic PC12 cell degeneration by regulating p-PKC-α/p-ERK1/2-Nurr1 signaling and inducing autophagy. VEGF-mediated angiogenesis was markedly enhanced in the conditioned media of 6-OHDA-lesioned PC12 cells with CYS C-overexpression, whereas blockage of autophagy in CYS C-overexpressing PC12 cells significantly downregulated VEGF expression and the associated angiogenesis. Our data indicate that CYS C displays dual neuronal-vascular functions, promoting PC12 cell survival and angiogenesis via regulating the level of secreted VEGF in NVUs. Our study provides evidence that may aid in the development of an alternative approach for the treatment of PD through modulation of CYS C-mediated neuronal-vascular pathways.


Asunto(s)
Cistatina C/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/genética , Factor A de Crecimiento Endotelial Vascular/genética , alfa-Sinucleína/genética , Animales , Autofagia/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inyecciones Intraventriculares , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Oxidopamina/antagonistas & inhibidores , Oxidopamina/farmacología , Células PC12 , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ratas , Transducción de Señal , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , alfa-Sinucleína/metabolismo
15.
J Leukoc Biol ; 101(5): 1233-1243, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28196851

RESUMEN

Inflammation-induced bone destruction is a major treatment target in many inflammatory skeletal diseases. The aim of this study was to investigate if the cysteine proteinase inhibitors cystatin C, fungal cysteine proteinase inhibitor (E-64), and N-benzyloxycarbonyl-arginyl-leucyl-valyl-glycyl-diazomethane acetate (Z-RLVG-CHN2) can inhibit LPS-induced osteoclast formation. Mouse bone marrow macrophages (BMMs) were isolated and primed with receptor activator of NF-κB ligand (RANKL) for 24 h, followed by stimulation with LPS, with and without inhibitors. Adult mice were injected locally with LPS and then treated with E-64 and osteoclast formation assessed by the number of cathepsin K+ multinucleated cells. Cystatin C inhibited LPS-induced osteoclast formation time and concentration dependently (IC50 = 0.3 µM). The effect was associated with decreased mRNA and protein expression of tartrate-resistant acid phosphatase (TRAP) and cathepsin K and of the osteoclastogenic transcription factors c-Fos and NFATc1. LPS-induced osteoclast formation on bone slices was also inhibited by cystatin C, resulting in decreased pit formation and release of bone matrix proteins. Similar data were obtained with E-64 and Z-RLVG-CHN2 Cystatin C was internalized in BMMs stimulated by LPS but not in unstimulated BMMs. Osteoclast formation induced by LPS was dependent on TNF-α, and the 3 inhibitors abolished LPS-induced TNF superfamily 2 (gene encoding TNF-α; Tnfsf2) mRNA expression without affecting Il1b, Il6, or oncostatin M (Osm) expression. Formation of osteoclasts in the skull bones after local LPS stimulation was inhibited by E-64. It is concluded that cysteine proteinase inhibitors effectively inhibit LPS-induced osteoclast formation in vivo and in vitro by inhibition of TNF-α expression. The targeting of cysteine proteinases might represent a novel treatment modality for prevention of inflammatory bone loss.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/prevención & control , Cistatina C/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Leucina/análogos & derivados , Oligopéptidos/farmacología , Osteoclastos/efectos de los fármacos , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Resorción Ósea/inducido químicamente , Resorción Ósea/genética , Resorción Ósea/inmunología , Catepsina K/genética , Catepsina K/inmunología , Relación Dosis-Respuesta Inmunológica , Regulación de la Expresión Génica , Interleucina-6/genética , Interleucina-6/inmunología , Leucina/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/inmunología , Oncostatina M/genética , Oncostatina M/inmunología , Osteoclastos/inmunología , Osteoclastos/patología , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/inmunología , Ligando RANK/antagonistas & inhibidores , Ligando RANK/farmacología , Transducción de Señal , Fosfatasa Ácida Tartratorresistente/genética , Fosfatasa Ácida Tartratorresistente/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
16.
Bioorg Med Chem ; 25(4): 1431-1439, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28110818

RESUMEN

A peptidomimetic called A20 (Cystapep 1) structurally based upon the N-terminal fragment of human cystatin C is known to have strong antibacterial properties. A20 is characterized by high activity against several bacterial strains often isolated from infected wounds, including methicillin-resistant S. aureus (MRSA). In this work we wanted to explore the therapeutic potential of A20 in the treatment of wound infections. We examined, cytotoxicity, allergenicity and impact of A20 on the proliferation and viability of human keratinocytes. Furthermore, the previously described antimicrobial action of A20has been confirmed here with reference strains of bacteria and extended by several other species. The A20 was highly active against Gram-positive bacteria with minimal inhibitory (MIC) and minimal bactericidal concentrations (MBC) between 8 and 128µg/mL. A20 did not affect proliferation of primary human keratinocytes in concentrations up to 50µg/mL. At the same time, it did not activate Peripheral Blood Mononuclear Cells (PBMCs), including basophils or neutrophils in vitro. Interestingly A20 was found to display immunomodulatory functions as it influences the production of Th2 cytokines (IL-4 and IL-13) by activated PBMCs. It was also resistant to degradation for at least 48h in human plasma. The results indicate that A20 is effective against the multiantibiotic-resistant bacteria and has a high safety profile, which makes it a promising antimicrobial drug candidate.


Asunto(s)
Antibacterianos/farmacología , Cistatina C/farmacología , Bacterias Grampositivas/efectos de los fármacos , Peptidomiméticos/farmacología , Infección de Heridas/tratamiento farmacológico , Antibacterianos/síntesis química , Antibacterianos/química , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Cistatina C/síntesis química , Cistatina C/química , Relación Dosis-Respuesta a Droga , Humanos , Queratinocitos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Relación Estructura-Actividad , Infección de Heridas/microbiología
17.
Ren Fail ; 38(9): 1496-1502, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27604130

RESUMEN

BACKGROUND: The aim of the current study was to investigate whether agmatine (AGM) has a protective effect against cisplatin-induced nephrotoxicity. MATERIALS AND METHODS: Thirty-two rats were randomly divided into four groups: (1) Saline (control); (2) Cisplatin (CDDP; 7.5 mg/kg intraperitoneally); (3) Agmatine (AGM; 10 mg/kg intraperitoneally); (4) Cisplatin plus agmatine (CDDP + AGM). Agmatine was given before and two consecutive days after cisplatin injection. All the animals underwent renal scintigraphy with 99mTc-DMSA. The levels of serum creatinine, cystatin C, and blood urea nitrogen (BUN) were measured in addition to examination of the tissue samples with light microscopy. Acute renal injury was assessed with biochemical analyses, scintigraphic imaging, and histopathological evaluation. RESULTS: In the cisplatin group, the levels of BUN, creatinine, and cystatin C were significantly higher than that of the controls. Histopathological examination showed remarkable damage of tubular and glomerular structures. Additionally, cisplatin caused markedly decreased renal 99mTc-DMSA uptake. AGM administration improved renal functions. Serum creatinine, BUN, and cystatin C levels had a tendency to normalize and, scintigraphic and histopathological findings showed significantly less evidence of renal toxicity than those observed in animals receiving cisplatin alone. CONCLUSIONS: Our data indicate that AGM has a protective effect against cisplatin-induced nephrotoxicity. Therefore, it may improve the therapeutic index of cisplatin. In addition, the early renal damage induced by cisplatin and protective effects of AGM against cisplatin nephrotoxicity was accurately demonstrated with 99mTc-DMSA renal scintigraphy.


Asunto(s)
Lesión Renal Aguda/prevención & control , Agmatina/farmacología , Cistatina C/farmacología , Riñón/diagnóstico por imagen , Cintigrafía/métodos , Ácido Dimercaptosuccínico de Tecnecio Tc 99m/farmacología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/diagnóstico , Animales , Cisplatino/toxicidad , Modelos Animales de Enfermedad , Femenino , Radiofármacos/farmacología , Ratas , Ratas Wistar
18.
Int J Biochem Cell Biol ; 79: 199-208, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27592448

RESUMEN

Cathepsins are powerful proteases, once referred to as the lysosomal cysteine proteases, that have been implicated in breast cancer invasion and metastasis, but pharmaceutical inhibitors have suffered failures in clinical trials due to adverse side effects. Scientific advancement from lysosomotropic to cell impermeable cathepsin inhibitors have improved efficacy in treating disease, but off-target effects have still been problematic, motivating a need to better understand cellular feedback and responses to treatment with cathepsin inhibitors. To address this need, we investigated effects of E-64 and cystatin C, two broad spectrum cathepsin inhibitors, on cathepsin levels intra- and extracellularly in MDA-MB-231 breast cancer cells. Cathepsins S and L had opposing responses to both E-64 and cystatin C inhibitor treatments with paradoxically elevated amounts of active cathepsin S, but decreased amounts of active cathepsin L, as determined by multiplex cathepsin zymography. This indicated cellular feedback to selectively sustain the amounts of active cathepsin S even in the presence of inhibitors with subnanomolar inhibitory constant values. These differences were identified in cellular locations of cathepsins L and S, trafficking for secretion, co-localization with endocytosed inhibitors, and longer protein turnover time for cathepsin S compared to cathepsin L. Together, this work demonstrates that previously underappreciated cellular compensation and compartmentalization mechanisms may sustain elevated amounts of some active cathepsins while diminishing others after inhibitor treatment. This can confound predictions based solely on inhibitor kinetics, and must be better understood to effectively deploy therapies and dosing strategies that target cathepsins to prevent cancer progression.


Asunto(s)
Neoplasias de la Mama/patología , Catepsina L/antagonistas & inhibidores , Catepsinas/antagonistas & inhibidores , Cistatina C/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Leucina/análogos & derivados , Inhibidores de Proteasas/farmacología , Catepsina L/metabolismo , Catepsinas/metabolismo , Línea Celular Tumoral , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Humanos , Leucina/farmacología , Transporte de Proteínas/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
19.
PLoS One ; 11(8): e0161093, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27532339

RESUMEN

Amyloid-ß (Aß), the major component of neuritic plaques in Alzheimer's disease (AD), is derived from sequential proteolytic cleavage of amyloid protein precursor (APP) by secretases. In this study, we found that cystatin C (CysC), a natural cysteine protease inhibitor, is able to reduce Aß40 secretion in human brain microvascular endothelial cells (HBMEC). The CysC-induced Aß40 reduction was caused by degradation of ß-secretase BACE1 through the ubiquitin/proteasome pathway. In contrast, we found that CysC promoted secretion of soluble APPα indicating the activated non-amyloidogenic processing of APP in HBMEC. Further results revealed that α-secretase ADAM10, which was transcriptionally upregulated in response to CysC, was required for the CysC-induced sAPPα secretion. Knockdown of SIRT1 abolished CysC-triggered ADAM10 upregulation and sAPPα production. Taken together, our results demonstrated that exogenously applied CysC can direct amyloidogenic APP processing to non-amyloidgenic pathway in brain endothelial cells, mediated by proteasomal degradation of BACE1 and SIRT1-mediated ADAM10 upregulation. Our study unveils previously unrecognized protective role of CysC in APP processing.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/efectos de los fármacos , Cistatina C/metabolismo , Cistatina C/farmacología , Fragmentos de Péptidos/biosíntesis , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Sustancias Protectoras/metabolismo , Sustancias Protectoras/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sirtuina 1/metabolismo , Regulación hacia Arriba/efectos de los fármacos
20.
J Leukoc Biol ; 100(4): 811-822, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27190304

RESUMEN

Human cystatin C, a member of the cysteine proteinase-inhibitory family, is produced by all nucleated cells and has important roles in regulating natural immunity. Nematode homologs to human cystatin C have been shown to have anti-inflammatory effects on monocytes and to reduce colitis in mice. In Crohn's disease, pathogenic activated monocytes help drive inflammatory processes via the release of proinflammatory cytokines and chemokines. In particular, tumor necrosis factor-α-producing inflammatory monocytes have a central role in the intestinal inflammation in patients with Crohn's disease. We investigated the potential of human cystatin C to regulate pathogenic activated monocytes and its potential as an Immunomodulator in Crohn's disease. We found that cystatin C significantly decreased the lipopolysaccharide-stimulated release and expression of interleukin-1ß and tumor necrosis factor-α in monocyte and peripheral blood mononuclear cell cultures from healthy donors, whereas interleukin-6 and interleukin-8 levels were unchanged. A similar reduction of interleukin-1ß and tumor necrosis factor-α was also seen in peripheral blood mononuclear cell cultures from patients with Crohn's disease, and in particular, tumor necrosis factor-α was reduced in supernatants from lamina propria cell cultures from patients with Crohn's disease. Further investigation revealed that cystatin C was internalized by monocytes via an active endocytic process, decreased phosphorylation of the mitogen-activated protein kinase pathway extracellular signal-regulated kinase-1/2, and altered surface marker expression. The ability of cystatin C to modulate the cytokine expression of monocytes, together with its protease-inhibitory function, indicates that modulation of the local cystatin C expression could be an option in future Crohn's disease therapy.


Asunto(s)
Enfermedad de Crohn/inmunología , Cistatina C/farmacología , Interleucina-1beta/metabolismo , Monocitos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/genética , Carbocianinas , Caspasa 1/biosíntesis , Caspasa 1/genética , Células Cultivadas , Enfermedad de Crohn/sangre , Cistatina C/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-6/análisis , Interleucina-8/análisis , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Monocitos/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...