Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Exp Nephrol ; 2024 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-38402502

RESUMEN

BACKGROUND: Volume overload is common and associated with high mortality in patients on peritoneal dialysis (PD). Traditional strategies including diuretics, water/salt restriction, and icodextrin-based solutions cannot always fully correct this condition, necessitating novel alternative strategies. Recent studies confirmed the expression of sodium-glucose cotransporter 2 (SGLT2) in the human peritoneum. Experimental data suggest that SGLT2 inhibitors decrease glucose absorption from the PD solution, thereby increasing the ultrafiltration volume. This trial aims to assess whether SGLT2 inhibitors increase the ultrafiltration volume in patients on PD. METHODS: The EMPOWERED trial (trial registration: jRCTs051230081) is a multicenter, randomized, double-blind, placebo-controlled, crossover trial. Patients with clinically diagnosed chronic heart failure are eligible regardless of the presence of diabetes if they use at least 3 L/day glucose-based PD solutions. Participants will be randomly assigned (1:1) to receive empagliflozin 10 mg once daily and then placebo or vice versa. Each treatment period will last 8 weeks with a 4-week washout period. This study will recruit at least 36 randomized participants. The primary endpoint is the change in the daily ultrafiltration volume from baseline to week 8 in each intervention period. The key secondary endpoints include changes in the biomarkers of drained PD solutions, renal residual function, and anemia-related parameters. CONCLUSIONS: This trial aims to assess the benefit of SGLT2 inhibitors in fluid management with a novel mechanism of action in patients on PD. It will also provide insights into the effects of SGLT2 inhibitors on solute transport across the peritoneal membrane and residual renal function.

2.
CEN Case Rep ; 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37962818

RESUMEN

Shiga toxin-producing Escherichia coli-associated hemolytic uremic syndrome (STEC-HUS) can induce life-threatening complications, including acute kidney injury, encephalopathy, and gastrointestinal complications. On the other hand, there have been few reports of cholecystitis associated with STEC-HUS. In this study, we report the case of an 83-year-old Japanese man who developed recurrent acute cholecystitis associated with STEC-HUS. Prior to establishing a definite diagnosis of STEC-HUS, plasma exchange and hemodialysis were initiated, which resulted in a rapid increase in the platelet count and decrease in lactate dehydrogenase levels. The patient presented an enlarged gallbladder detected by computed tomography during the course of treatment. Due to recurrent flare-ups, the patient had to undergo several rounds of endoscopic retrograde biliary drainage and, ultimately, cholecystectomy to prevent relapse of acute cholecystitis. Since cholecystitis was thought to have been caused by complex mechanisms in this case, we discussed those from multiple perspectives. This case report highlights the need for particular care to be given to the management of pre-existing diseases as well as STEC-HUS, especially in older patients.

3.
Intern Med ; 2023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37779073

RESUMEN

Pregnancies with chronic kidney disease (CKD) and high disease activity in rheumatic diseases are high-risk events with adverse outcomes for both the mother and fetus. We herein report a 35-year-old woman with juvenile idiopathic arthritis (JIA), amyloid A (AA) amyloidosis related to JIA, and CKD stage G4A2 who wished to have children. She achieved a successful pregnancy, even in the presence of these multiple risk factors, using tocilizumab to control the disease activity of JIA and AA amyloidosis, along with antihypertensive drugs to control her blood pressure before and during pregnancy.

5.
PLoS One ; 18(2): e0282163, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36827357

RESUMEN

BACKGROUND: Management of diabetic kidney disease (DKD) to prevent end-stage kidney disease (ESKD) has become a major challenge for health care professionals. This study aims to investigate the characteristics of patients with DKD when they are first referred to a nephrologist and the subsequent prognoses. METHODS: A total of 307 patients who were referred to our department from October 2010 to September 2014 at Osaka General Medical Center were analyzed. Independent risk factors associated with renal replacement therapy (RRT) and cardiovascular composite events (CVE) following their nephrology referral were later identified using Cox proportional hazards analysis. RESULTS: Of 307 patients, 26 (8.5%), 67 (21.8%), 134 (43.6%), and 80 (26.1%) patients were categorized as having chronic kidney disease (CKD) stages 3a, 3b, 4, and 5, respectively. The median estimated glomerular filtration rate (eGFR) and urinary protein levels were 22.3 mL/min/1.73 m2 and 2.83 g/gCr, respectively, at the time of the nephrology referral. During the follow-up period (median, 30 months), 121 patients required RRT, and more than half of the patients with CKD stages 5 and 4 reached ESKD within 60 months following their nephrology referral; 30% and <10% of the patients with CKD stages 3b and 3a, respectively, required RRT within 60 months following their nephrology referral. CONCLUSION: Patients with DKD were referred to nephrologist at CKD stage 4. Although almost half of the patients with CKD stage 5 at the time of nephrology referral required RRT within one-and-a-half years after the referral, kidney function of patients who were referred to nephrologist at CKD stage 3 and 4 were well preserved.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Fallo Renal Crónico , Nefrología , Insuficiencia Renal Crónica , Humanos , Nefropatías Diabéticas/complicaciones , Nefrólogos , Estudios Retrospectivos , Progresión de la Enfermedad , Fallo Renal Crónico/terapia , Insuficiencia Renal Crónica/complicaciones , Pronóstico , Tasa de Filtración Glomerular , Derivación y Consulta
6.
Clin Exp Nephrol ; 24(8): 705-714, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32281008

RESUMEN

BACKGROUND: The duration of predialysis nephrological care that can reduce all-cause and cardiovascular mortality after dialysis initiation has not been clarified. METHODS: A total of 1117 patients who started chronic dialysis treatment from 2006 to 2015 at Osaka General Medical Center were analyzed. Independent risk factors associated with all-cause and cardiovascular mortality after dialysis initiation and early death (death within 12 months after dialysis initiation) were identified using Cox proportional hazards analysis. Moreover, the duration of predialysis nephrology care that could reduce mortality was explored using several different definitions of early referral as well as "6 months" commonly used in previous studies. RESULTS: Of 1117 patients, 834 were referred 6 months before dialysis initiation. During the follow-up period (median, 34 months), 324 patients died after dialysis initiation. Although multivariate Cox analysis did not show a favorable association between early referral of "6 months before dialysis initiation" and all-cause and cardiovascular mortality, 20-month predialysis nephrological care was associated with better first-year overall survival after dialysis initiation (hazard ratio 0.58; 95% confidence interval 0.35-0.98; P = 0.040). CONCLUSION: More than 6 months nephrological care before dialysis initiation was not early enough to reduce all-cause and cardiovascular mortality after dialysis initiation. Our results suggest that nephrology referral 20 months before dialysis initiation would be necessary to reduce first-year overall survival after dialysis initiation.


Asunto(s)
Fallo Renal Crónico/terapia , Anciano , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/mortalidad , Femenino , Humanos , Japón/epidemiología , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/mortalidad , Masculino , Persona de Mediana Edad , Diálisis Renal , Estudios Retrospectivos , Factores de Tiempo
7.
Kidney Int ; 96(1): 67-79, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30910380

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic microangiopathy (TMA) caused by dysregulated complement activation. Clinically, aHUS is effectively treated by an anti-C5 monoclonal antibody (mAb) but whether the disease is mediated by the C5a receptor (C5aR) or C5b-9 pathway, or both, is unknown. Here we address this in a factor H mutant mouse (FHR/R) which developed complement-mediated TMA as well as macrovascular thrombosis caused by an aHUS-related factor H point mutation (mouse W1206R, corresponding to human W1183R). C5 deficiency and anti-C5 mAb treatment blocked all disease manifestations in FHR/R mice. C5aR1 gene deficiency prevented macrovascular thrombosis in various organs but did not improve survival or reduce renal TMA. Conversely, C6 or C9 deficiency significantly improved survival and markedly diminished renal TMA but did not prevent macrovascular thrombosis. Interestingly, as they aged both FHR/R C6-/- and FHR/R C9-/- mice developed glomerular disease reminiscent of C3 glomerulonephritis. Thus, C5aR and C5b-9 pathways drove different aspects of disease in FHR/R mice with the C5aR pathway being responsible for macrovascular thrombosis and chronic inflammatory injury while the C5b-9 pathway caused renal TMA. Our data provide new understanding of the pathogenesis of complement-mediated TMA and macrovascular thrombosis in FHR/R mice and suggest that C5 blockade is more effective for the treatment of aHUS than selectively targeting the C5aR or C5b-9 pathway alone.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/inmunología , Factor H de Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Glomérulos Renales/patología , Receptor de Anafilatoxina C5a/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/patología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C6/genética , Complemento C6/inmunología , Complemento C6/metabolismo , Factor H de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/ultraestructura , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica , Mutación Puntual , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo
8.
Am J Pathol ; 189(4): 826-838, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30711487

RESUMEN

Single-nucleotide polymorphisms and rare mutations in factor H (FH; official name, CFH) are associated with age-related macular degeneration and atypical hemolytic uremic syndrome, a form of thrombotic microangiopathy. Mice with the FH W1206R mutation (FHR/R) share features with human atypical hemolytic uremic syndrome. Herein, we report that FHR/R mice exhibited retinal vascular occlusion and ischemia. Retinal fluorescein angiography demonstrated delayed perfusion and vascular leakage in FHR/R mice. Optical coherence tomography imaging of FHR/R mice showed retinal degeneration, edema, and detachment. Histologic analysis of FHR/R mice revealed retinal thinning, vessel occlusion, as well as degeneration of photoreceptors and retinal pigment epithelium. Immunofluorescence showed albumin leakage from blood vessels into the neural retina, and electron microscopy demonstrated vascular endothelial cell irregularity with narrowing of retinal and choroidal vessels. Knockout of C6, a component of the membrane attack complex, prevented the aforementioned retinal phenotype in FHR/R mice, consistent with membrane attack complex-mediated pathogenesis. Pharmacologic blockade of C5 also rescued retinas of FHR/R mice. This FHR/R mouse strain represents a model for retinal vascular occlusive disorders and ischemic retinopathy. The results suggest complement dysregulation can contribute to retinal vascular occlusion and that an anti-C5 antibody might be helpful for C5-mediated thrombotic retinal diseases.


Asunto(s)
Factor H de Complemento/fisiología , Isquemia/etiología , Mutación , Neovascularización Patológica/etiología , Enfermedades de la Retina/etiología , Epitelio Pigmentado de la Retina/patología , Trombosis/etiología , Animales , Factor H de Complemento/genética , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Epitelio Pigmentado de la Retina/metabolismo , Trombosis/metabolismo , Trombosis/patología
9.
Intern Med ; 57(24): 3603-3610, 2018 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-30101934

RESUMEN

We present a case of Dent disease caused by a novel intronic mutation, 1348-1G>A, of the chloride voltage-gated channel 5 (CLCN5) gene. Cultured proximal tubule cells obtained from the patient showed impaired acidification of the endosome and/or lysosome, indicating that the 1348-1G>A mutation was indeed the cause of Dent disease. Although the prevalence of osteomalacia in Dent disease is low in Japan, several factors-including poor medication adherence-caused severe osteomalacia in the current case. Oral supplementation with calcium and native/active vitamin D therapy, with careful attention to medication adherence, led to the improvement of the patient's bone status.


Asunto(s)
Canales de Cloruro/genética , Enfermedad de Dent/genética , Osteomalacia/genética , Mutación Puntual , Adulto , Calcio de la Dieta/uso terapéutico , Enfermedad de Dent/complicaciones , Enfermedad de Dent/patología , Suplementos Dietéticos , Humanos , Intrones , Japón , Túbulos Renales Proximales/patología , Masculino , Cumplimiento de la Medicación , Osteomalacia/tratamiento farmacológico , Osteomalacia/etiología , Osteomalacia/patología , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico
10.
J Am Soc Nephrol ; 29(8): 2053-2059, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29895552

RESUMEN

Background C3 glomerulopathy (C3G) is a life-threatening kidney disease caused by dysregulation of the alternative pathway of complement (AP) activation. No approved specific therapy is available for C3G, although an anti-C5 mAb has been used off-label in some patients with C3G, with mixed results. Thus, there is an unmet medical need to develop other inhibitors of complement for C3G.Methods We used a murine model of lethal C3G to test the potential efficacy of an Fc fusion protein of complement receptor of the Ig superfamily (CRIg-Fc) in the treatment of C3G. CRIg-Fc binds C3b and inhibits C3 and C5 convertases of the AP. Mice with mutations in the factor H and properdin genes (FHm/mP-/-) develop early-onset C3G, with AP consumption, high proteinuria, and lethal crescentic GN.Results Treatment of FHm/mP-/- mice with CRIg-Fc, but not a control IgG, inhibited AP activation and diminished the consumption of plasma C3, factor B, and C5. CRIg-Fc-treated FHm/mP-/- mice also had significantly improved survival and reduced proteinuria, hematuria, BUN, glomerular C3 fragment, C9 and fibrin deposition, and GN pathology scores.Conclusions Therapeutics developed on the basis of the mechanism of action of soluble CRIg may be effective for the treatment of C3G and should be explored clinically.


Asunto(s)
Complemento C3/antagonistas & inhibidores , Complemento C3/genética , Glomerulonefritis por IGA/genética , Glomerulonefritis por IGA/prevención & control , Factor 2 Liberador de Guanina Nucleótido/genética , Receptores de Complemento/genética , Análisis de Varianza , Animales , Biopsia con Aguja , Western Blotting , Activación de Complemento , Factor B del Complemento/inmunología , Factor B del Complemento/metabolismo , Modelos Animales de Enfermedad , Glomerulonefritis por IGA/patología , Inmunohistoquímica , Pruebas de Función Renal , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Receptores de Complemento/metabolismo , Tasa de Supervivencia
11.
J Immunol ; 201(3): 1021-1029, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29898960

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is a serious blood disorder characterized by dysregulated complement activation on blood cells. Eculizumab, the current standard therapy and a humanized anti-C5 mAb, relieves anemia and thrombosis symptoms of PNH patients by preventing complement-dependent intravascular hemolysis (IVH). However, up to 20% of PNH patients on long-term eculizumab treatment still suffer from significant anemia and are transfusion dependent because of extravascular hemolysis (EVH) of C3-opsonized PNH erythrocytes. In this study, we show that function-blocking anti-properdin (P) mAbs dose-dependently inhibited autologous, complement-mediated hemolysis induced by factor H dysfunction. Furthermore, anti-human P (hP) mAbs potently and dose-dependently inhibited acidified serum-induced hemolysis of PNH erythrocytes (Ham test). In contrast to erythrocytes rescued by anti-C5 mAb, nonlysed PNH erythrocytes rescued by anti-P mAb incurred no activated C3 fragment deposition on their surface. These results suggested that anti-P mAbs may prevent EVH as well as IVH of PNH erythrocytes. To test the in vivo efficacy of anti-hP mAbs in preventing EVH, we generated a P humanized mouse by transgenic expression of hP in P knockout mice (hP-Tg/P-/-). In a murine EVH model, complement-susceptible erythrocytes were completely eliminated within 3 d in control mAb-treated hP-Tg/P-/- mice, whereas such cells were protected and persisted in hP-Tg/P-/- mice treated with an anti-hP mAb. Collectively, these data suggest that anti-P mAbs can inhibit both IVH and EVH mediated by complement and may offer improved efficacy over eculizumab, the current standard therapy for PNH.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Activación de Complemento/inmunología , Hemólisis/inmunología , Properdina/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Eritrocitos/inmunología , Femenino , Hemoglobinuria Paroxística/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
J Am Soc Nephrol ; 29(7): 1928-1937, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29858280

RESUMEN

Background Properdin (P) is a positive regulator of the alternative pathway of complement activation. Although P inhibition is expected and has been shown to ameliorate the alternative pathway of complement-mediated tissue injury in several disease models, it unexpectedly exacerbated renal injury in a murine model of C3 glomerulopathy. The role of P in atypical hemolytic uremic syndrome (aHUS) is uncertain.Methods We blocked P function by genetic deletion or mAb-mediated inhibition in mice carrying a factor H (FH) point mutation, W1206R (FHR/R), that causes aHUS and systemic thrombophilia with high mortality.Results P deficiency completely rescued FHR/R mice from premature death and prevented thrombocytopenia, hemolytic anemia, and renal disease. It also eliminated macrovessel thrombi that were prevalent in FHR/R mice. All mice that received a function-blocking anti-P mAb for 8 weeks survived the experimental period and appeared grossly healthy. Platelet counts and hemoglobin levels were significantly improved in FHR/R mice after 4 weeks of anti-P mAb treatment. One half of the FHR/R mice treated with an isotype control mAb but none of the anti-P mAb-treated mice developed stroke-related neurologic disease. Anti-P mAb-treated FHR/R mice showed largely normal renal histology, and residual liver thrombi were detected in only three of 15 treated mice.Conclusions These results contrast with the detrimental effect of P inhibition observed in a murine model of C3 glomerulopathy and suggest that P contributes critically to aHUS pathogenesis. Inhibition of P in aHUS may be of therapeutic benefit.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Complemento C3/metabolismo , Complemento C9/metabolismo , Properdina/genética , Trombofilia/genética , Animales , Anticuerpos Monoclonales/uso terapéutico , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/prevención & control , Factor H de Complemento/genética , Vía Alternativa del Complemento , Femenino , Fibrina/metabolismo , Hemoglobinas/metabolismo , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Recuento de Plaquetas , Properdina/deficiencia , Properdina/inmunología , Trombofilia/prevención & control , Trombosis/prevención & control
13.
Kidney Int ; 91(6): 1386-1397, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28139294

RESUMEN

C3 glomerulopathy is a potentially life-threatening disease of the kidney caused by dysregulated alternative pathway complement activation. The specific complement mediator(s) responsible for kidney injury in C3 glomerulopathy are yet to be defined and no specific therapy is currently available. We previously developed a mouse model of lethal C3 glomerulopathy with factor H and properdin gene double mutations. Therefore, we used this model to examine the role of C5 and C5a receptor (C5aR) in the pathogenesis of the disease. Disease severity in these factor H/properdin double-mutant mice was found to be correlated with plasma C5 levels, and prophylactic anti-C5 mAb therapy was effective in preventing lethal C3 glomerulopathy. When given to these double-mutant mice that had already developed active disease with severe proteinuria, anti-C5 mAb treatment also prevented death in half of the mice. Deficiency of C5aR significantly reduced disease severity, suggesting that C5aR-mediated inflammation contributed to C3 glomerulopathy. Thus, C5 and C5aR have a critical role in C3 glomerulopathy. Hence, early intervention targeting these pathways may be an effective therapeutic strategy for patients with C3 glomerulopathy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Complemento C3/metabolismo , Complemento C5/antagonistas & inhibidores , Inactivadores del Complemento/farmacología , Vía Alternativa del Complemento/efectos de los fármacos , Glomerulonefritis/prevención & control , Riñón/efectos de los fármacos , Insuficiencia Renal/prevención & control , Animales , Complemento C3/inmunología , Complemento C5/inmunología , Complemento C5/metabolismo , Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Glomerulonefritis/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Properdina/deficiencia , Properdina/genética , Proteinuria/inmunología , Proteinuria/metabolismo , Proteinuria/prevención & control , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Insuficiencia Renal/genética , Insuficiencia Renal/inmunología , Insuficiencia Renal/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
14.
Blood ; 129(9): 1184-1196, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28057640

RESUMEN

Complement plays a key role in host defense, but its dysregulation can cause autologous tissue injury. Complement activation is normally controlled by regulatory proteins, including factor H (FH) in plasma and membrane cofactor protein (MCP) on the cell surface. Mutations in FH and MCP are linked to atypical hemolytic uremic syndrome, a type of thrombotic microangiopathy (TMA) that causes renal failure. We describe here that disruption of FH function on the cell surface can also lead to disseminated complement-dependent macrovascular thrombosis. By gene targeting, we introduced a point mutation (W1206R) into murine FH that impaired its interaction with host cells but did not affect its plasma complement-regulating activity. Homozygous mutant mice carrying this mutation developed renal TMA as well as systemic thrombophilia involving large blood vessels in multiple organs, including liver, lung, spleen, and kidney. Approximately 30% of mutant mice displayed symptoms of stroke and ischemic retinopathy, and 48% died prematurely. Genetic deficiency of complement C3 and factor D prevented both the systemic thrombophilia and renal TMA phenotypes. These results demonstrate a causal relationship between complement dysregulation and systemic angiopathy and suggest that complement activation may contribute to various human thrombotic disorders involving both the micro- and macrovasculature.


Asunto(s)
Factor H de Complemento/genética , Síndrome Hemolítico-Urémico/genética , Trombofilia/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Puntual
15.
Immunobiology ; 221(10): 1080-90, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27371974

RESUMEN

Remarkable advances have been made over past decades in understanding the pathogenesis of complement-mediated diseases. This has led to development of new therapies for, and in some cases re-classification of, complement-driven diseases. This success is due to not only insight from human patients but also studies using transgenic animal models. Animal models that mimic human diseases are useful tools to understand the mechanism of disease and develop new therapies but there are also limitations due to species differences in their complement systems. This review provides a summary of transgenic animal models for three human diseases that are at the forefront of anti-complement therapy, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G). They are discussed here as examples to highlight the values and limitations of animal modeling in complement-driven diseases.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Susceptibilidad a Enfermedades , Animales , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/etiología , Síndrome Hemolítico Urémico Atípico/metabolismo , Activación de Complemento/inmunología , Inactivadores del Complemento/farmacología , Inactivadores del Complemento/uso terapéutico , Modelos Animales de Enfermedad , Hemoglobinuria Paroxística/tratamiento farmacológico , Hemoglobinuria Paroxística/etiología , Hemoglobinuria Paroxística/metabolismo , Humanos , Ratones Transgénicos , Nefrosis/tratamiento farmacológico , Nefrosis/etiología , Nefrosis/metabolismo
16.
Nature ; 532(7597): 117-21, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27027293

RESUMEN

Colonic epithelial cells are covered by thick inner and outer mucus layers. The inner mucus layer is free of commensal microbiota, which contributes to the maintenance of gut homeostasis. In the small intestine, molecules critical for prevention of bacterial invasion into epithelia such as Paneth-cell-derived anti-microbial peptides and regenerating islet-derived 3 (RegIII) family proteins have been identified. Although there are mucus layers providing physical barriers against the large number of microbiota present in the large intestine, the mechanisms that separate bacteria and colonic epithelia are not fully elucidated. Here we show that Ly6/PLAUR domain containing 8 (Lypd8) protein prevents flagellated microbiota invading the colonic epithelia in mice. Lypd8, selectively expressed in epithelial cells at the uppermost layer of the large intestinal gland, was secreted into the lumen and bound flagellated bacteria including Proteus mirabilis. In the absence of Lypd8, bacteria were present in the inner mucus layer and many flagellated bacteria invaded epithelia. Lypd8(-/-) mice were highly sensitive to intestinal inflammation induced by dextran sulfate sodium (DSS). Antibiotic elimination of Gram-negative flagellated bacteria restored the bacterial-free state of the inner mucus layer and ameliorated DSS-induced intestinal inflammation in Lypd8(-/-) mice. Lypd8 bound to flagella and suppressed motility of flagellated bacteria. Thus, Lypd8 mediates segregation of intestinal bacteria and epithelial cells in the colon to preserve intestinal homeostasis.


Asunto(s)
Colon/microbiología , Epitelio/microbiología , Flagelos , Proteínas Ligadas a GPI/metabolismo , Bacterias Gramnegativas/fisiología , Mucosa Intestinal/microbiología , Animales , Adhesión Bacteriana , Células CACO-2 , Línea Celular , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/genética , Sulfato de Dextran , Femenino , Proteínas Ligadas a GPI/deficiencia , Proteínas Ligadas a GPI/genética , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/metabolismo , Bacterias Gramnegativas/patogenicidad , Homeostasis , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/genética , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Masculino , Ratones , Proteus mirabilis/efectos de los fármacos , Proteus mirabilis/metabolismo , Proteus mirabilis/patogenicidad , Simbiosis
17.
PLoS One ; 8(2): e55800, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23409047

RESUMEN

Although the pyrazolone derivative sulpyrine is widely used as an antipyretic analgesic drug, side effects, including fatal shock, have been reported. However, the molecular mechanism underlying such a severe side effect is largely unclear. Here, we report that the transcription factor CREBH that is highly expressed in the liver plays an important role in fatal shock induced by sulpyrine in mice. CREBH-deficient mice were resistant to experimental fatal sulpyrine shock. We found that sulpyrine-induced expression of cytochrome P450 2B (CYP2B) family genes, which are involved in sulpyrine metabolism, in the liver was severely impaired in CREBH-deficient mice. Moreover, introduction of CYP2B in CREBH-deficient liver restored susceptibility to sulpyrine. Furthermore, ectopic expression of CREBH up-regulated CYP2B10 promoter activity, and in vivo knockdown of CREBH in wild-type mice conferred a significant resistance to fatal sulpyrine shock. These data demonstrate that CREBH is a positive regulator of CYP2B in response to sulpyrine administration, which possibly results in fatal shock.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Dipirona/efectos adversos , Choque/inducido químicamente , Choque/genética , Ampirona/sangre , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/deficiencia , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citocromo P-450 CYP2B1/genética , Citocromo P-450 CYP2B1/metabolismo , Familia 2 del Citocromo P450 , Dipirona/farmacocinética , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Interferencia de ARN , Índice de Severidad de la Enfermedad , Choque/mortalidad , Esteroide Hidroxilasas/genética , Activación Transcripcional
18.
J Immunol ; 190(2): 774-83, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23241884

RESUMEN

Extracellular ATP is released from live cells in controlled conditions, as well as dying cells in inflammatory conditions, and, thereby, regulates T cell responses, including Th17 cell induction. The level of extracellular ATP is closely regulated by ATP hydrolyzing enzymes, such as ecto-nucleoside triphosphate diphosphohydrolases (ENTPDases). ENTPDase1/CD39, which is expressed in immune cells, was shown to regulate immune responses by downregulating the ATP level. In this study, we analyzed the immunomodulatory function of ENTPDase7, which is preferentially expressed in epithelial cells in the small intestine. The targeted deletion of Entpd7 encoding ENTPDase7 in mice resulted in increased ATP levels in the small intestinal lumen. The number of Th17 cells was selectively increased in the small intestinal lamina propria in Entpd7(-/-) mice. Th17 cells were decreased by oral administration of antibiotics or the ATP antagonist in Entpd7(-/-) mice, indicating that commensal microbiota-dependent ATP release mediates the enhanced Th17 cell development in the small intestinal lamina propria of Entpd7(-/-) mice. In accordance with the increased number of small intestinal Th17 cells, Entpd7(-/-) mice were resistant to oral infection with Citrobacter rodentium. Entpd7(-/-) mice suffered from severe experimental autoimmune encephalomyelitis, which was associated with increased numbers of CD4(+) T cells producing both IL-17 and IFN-γ. Taken together, these findings demonstrate that ENTPDase7 controls the luminal ATP level and, thereby, regulates Th17 cell development in the small intestine.


Asunto(s)
Adenosina Trifosfato/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Pirofosfatasas/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Citrobacter rodentium/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Femenino , Regulación de la Expresión Génica , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestino Delgado/microbiología , Metagenoma , Ratones , Ratones Noqueados , Pirofosfatasas/genética
19.
PLoS Pathog ; 8(5): e1002714, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693446

RESUMEN

Specific intestinal microbiota has been shown to induce Foxp3(+) regulatory T cell development. However, it remains unclear how development of another regulatory T cell subset, Tr1 cells, is regulated in the intestine. Here, we analyzed the role of two probiotic strains of intestinal bacteria, Lactobacillus casei and Bifidobacterium breve in T cell development in the intestine. B. breve, but not L. casei, induced development of IL-10-producing Tr1 cells that express cMaf, IL-21, and Ahr in the large intestine. Intestinal CD103(+) dendritic cells (DCs) mediated B. breve-induced development of IL-10-producing T cells. CD103(+) DCs from Il10(-/-), Tlr2(-/-), and Myd88(-/-) mice showed defective B. breve-induced Tr1 cell development. B. breve-treated CD103(+) DCs failed to induce IL-10 production from co-cultured Il27ra(-/-) T cells. B. breve treatment of Tlr2(-/-) mice did not increase IL-10-producing T cells in the colonic lamina propria. Thus, B. breve activates intestinal CD103(+) DCs to produce IL-10 and IL-27 via the TLR2/MyD88 pathway thereby inducing IL-10-producing Tr1 cells in the large intestine. Oral B. breve administration ameliorated colitis in immunocompromised mice given naïve CD4(+) T cells from wild-type mice, but not Il10(-/-) mice. These findings demonstrate that B. breve prevents intestinal inflammation through the induction of intestinal IL-10-producing Tr1 cells.


Asunto(s)
Bifidobacterium/inmunología , Colon/microbiología , Interleucina-10/metabolismo , Lacticaseibacillus casei/inmunología , Probióticos/administración & dosificación , Linfocitos T Reguladores/citología , Traslado Adoptivo , Animales , Infecciones por Bifidobacteriales/inmunología , Infecciones por Bifidobacteriales/microbiología , Infecciones por Bifidobacteriales/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular , Técnicas de Cocultivo , Colitis/inmunología , Colitis/microbiología , Colitis/terapia , Colon/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interacciones Huésped-Patógeno , Huésped Inmunocomprometido , Interleucina-10/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
20.
Proc Natl Acad Sci U S A ; 109(13): 5010-5, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22403066

RESUMEN

Adequate activation of CD4(+) T lymphocytes is essential for host defense against invading pathogens; however, exaggerated activity of effector CD4(+) T cells induces tissue damage, leading to inflammatory disorders such as inflammatory bowel diseases. Several unique subsets of intestinal innate immune cells have been identified. However, the direct involvement of innate immune cell subsets in the suppression of T-cell-dependent intestinal inflammation is poorly understood. Here, we report that intestinal CX(3)C chemokine receptor 1(high) (CX(3)CR1(high)) CD11b(+) CD11c(+) cells are responsible for prevention of intestinal inflammation through inhibition of T-cell responses. These cells inhibit CD4(+) T-cell proliferation in a cell contact-dependent manner and prevent T-cell-dependent colitis. The suppressive activity is abrogated in the absence of the IL-10/Stat3 pathway. These cells inhibit T-cell proliferation by two steps. Initially, CX(3)CR1(high) CD11b(+) CD11c(+) cells preferentially interact with T cells through highly expressed intercellular adhesion molecule-1/vascular cell adhesion molecule-1; then, they fail to activate T cells because of defective expression of CD80/CD86. The IL-10/Stat3 pathway mediates the reduction of CD80/CD86 expression. Transfer of wild-type CX(3)CR1(high) CD11b(+) CD11c(+) cells prevents development of colitis in myeloid-specific Stat3-deficient mice. Thus, these cells are regulatory myeloid cells that are responsible for maintaining intestinal homeostasis.


Asunto(s)
Colitis/inmunología , Colitis/patología , Intestinos/inmunología , Intestinos/patología , Células Mieloides/inmunología , Receptores de Quimiocina/metabolismo , Linfocitos T/inmunología , Animales , Antígenos CD11/metabolismo , Receptor 1 de Quimiocinas CX3C , Proliferación Celular , Colitis/complicaciones , Colitis/prevención & control , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Interleucina-10/metabolismo , Ratones , Membrana Mucosa/inmunología , Membrana Mucosa/patología , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...