Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Immunol ; 51(9): 2225-2236, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34146342

RESUMEN

Polymorphisms in TACI, a BAFF family cytokine receptor, are linked to diverse human immune disorders including common variable immunodeficiency (CVID) and systemic lupus erythematosus (SLE). Functional studies of individual variants show modest impacts on surface TACI expression and/or downstream signal transduction, indicating that relatively subtle variation in TACI activity can impact human B-cell biology. However, significant complexity underlies TACI biology, including both positive and negative regulation of physiologic and pathogenic B-cell responses. To model these contradictory events, we compared the functional impact of TACI deletion on separate models of murine SLE driven by T cell-independent and -dependent breaks in B-cell tolerance. First, we studied whether reduced surface TACI expression was sufficient to protect against progressive BAFF-mediated systemic autoimmunity. Strikingly, despite a relatively modest impact on surface TACI levels, TACI haploinsufficiency markedly reduced pathogenic RNA-associated autoantibody titers and conferred long-term protection from BAFF-driven lupus nephritis. In contrast, B cell-intrinsic TACI deletion exerted a limited impact of autoantibody generation in murine lupus characterized by spontaneous germinal center formation and T cell-dependent humoral autoimmunity. Together, these combined data provide new insights into TACI biology and highlight how TACI signals must be tightly regulated during protective and pathogenic B-cell responses.


Asunto(s)
Autoinmunidad/genética , Factor Activador de Células B/inmunología , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Autoinmunidad/inmunología , Factor Activador de Células B/antagonistas & inhibidores , Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/genética , Linfocitos B/inmunología , Quimera , Femenino , Haploinsuficiencia/genética , Inmunosupresores/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología
2.
Nephrol Dial Transplant ; 37(5): 847-859, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-34865099

RESUMEN

BACKGROUND: The sodium-glucose cotransporter-2 (SGLT2) inhibitor empagliflozin lowers blood glucose via reduced tubular reabsorption of filtered glucose and is an important new therapy for diabetic nephropathy (DN). This study tested whether treatment with empagliflozin would ameliorate proteinuria and the pathologic alterations of DN including podocyte number and integrity in the leptin-deficient BTBR ob/ob mouse model of DN. METHODS: Study cohorts included wild-type (WT) BTBR mice, untreated diabetic BTBR ob/ob mice and mice treated with empagliflozin for 6 weeks after development of established DN at 18 weeks of age. RESULTS: Hyperglycemia, proteinuria, serum creatinine, accumulation of mesangial matrix and the extent of mesangiolysis were reversed with empagliflozin treatment. Treatment with empagliflozin resulted in an increased podocyte number and podocyte density, improvement in the degree of podocyte foot process effacement and parietal epithelial cell activation. SGLT2 inhibition reduced renal oxidative stress, measured by urinary excretion of markers of RNA/DNA damage and in situ demonstration of decreased carbonyl oxidation. There was no discernable difference in accumulations of advanced glycation end-products by immunohistochemistry. CONCLUSION: The structural improvements seen in BTBR ob/ob mice treated with empagliflozin provide insights into potential long-term benefits for humans with DN, for whom there is no comparable biopsy information to identify structural changes effected by SGLT2 inhibition. The findings suggest SGLT2 inhibition may ameliorate DN through glucose lowering-dependent and -independent mechanisms that lead to podocyte restoration and delay or reversal of disease progress.


Asunto(s)
Compuestos de Bencidrilo , Diabetes Mellitus , Nefropatías Diabéticas , Glucósidos , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Compuestos de Bencidrilo/uso terapéutico , Glucemia , Diabetes Mellitus/tratamiento farmacológico , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/patología , Glucósidos/uso terapéutico , Ratones , Ratones Endogámicos , Proteinuria , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
3.
J Immunol ; 204(10): 2627-2640, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32238460

RESUMEN

Lupus nephritis (LN) is a major contributor to morbidity and mortality in lupus patients, but the mechanisms of kidney damage remain unclear. In this study, we introduce, to our knowledge, novel models of LN designed to resemble the polygenic nature of human lupus by embodying three key genetic alterations: the Sle1 interval leading to anti-chromatin autoantibodies; Mfge8-/- , leading to defective clearance of apoptotic cells; and either C1q-/- or C3-/- , leading to low complement levels. We report that proliferative glomerulonephritis arose only in the presence of all three abnormalities (i.e., in Sle1.Mfge8 -/- C1q -/- and Sle1.Mfge8 -/- C3 -/- triple-mutant [TM] strains [C1q -/-TM and C3-/- TM, respectively]), with structural kidney changes resembling those in LN patients. Unexpectedly, both TM strains had significant increases in autoantibody titers, Ag spread, and IgG deposition in the kidneys. Despite the early complement component deficiencies, we observed assembly of the pathogenic terminal complement membrane attack complex in both TM strains. In C1q-/- TM mice, colocalization of MASP-2 and C3 in both the glomeruli and tubules indicated that the lectin pathway likely contributed to complement activation and tissue injury in this strain. Interestingly, enhanced thrombin activation in C3-/- TM mice and reduction of kidney injury following attenuation of thrombin generation by argatroban in a serum-transfer nephrotoxic model identified thrombin as a surrogate pathway for complement activation in C3-deficient mice. These novel mouse models of human lupus inform the requirements for nephritis and provide targets for intervention.


Asunto(s)
Enfermedades por Deficiencia de Complemento Hereditario/genética , Riñón/patología , Nefritis Lúpica/inmunología , Animales , Anticuerpos Antinucleares/sangre , Antígenos de Superficie/genética , Activación de Complemento , Complemento C1q/genética , Complemento C3/genética , Modelos Animales de Enfermedad , Glomerulonefritis , Enfermedades por Deficiencia de Complemento Hereditario/inmunología , Humanos , Nefritis Lúpica/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Leche/genética , Herencia Multifactorial
4.
Lab Invest ; 101(7): 935-941, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33911188

RESUMEN

The etiology of diabetic nephropathy in type 2 diabetes is multifactorial. Sustained hyperglycemia is a major contributor, but additional contributions come from the hypertension, obesity, and hyperlipidemia that are also commonly present in patients with type 2 diabetes and nephropathy. The leptin deficient BTBR ob/ob mouse is a model of type 2 diabetic nephropathy in which hyperglycemia, obesity, and hyperlipidemia, but not hypertension, are present. We have shown that reversal of the constellation of these metabolic abnormalities with leptin replacement can reverse the morphologic and functional manifestations of diabetic nephropathy. Here we tested the hypothesis that reversal specifically of the hypertriglyceridemia, using an antisense oligonucleotide directed against ApoC-III, an apolipoprotein that regulates the interactions of VLDL (very low density lipoproteins) with the LDL receptor, is sufficient to ameliorate the nephropathy of Type 2 diabetes. Antisense treatment resulted in reduction of circulating ApoC-III protein levels and resulted in substantial lowering of triglycerides to near-normal levels in diabetic mice versus controls. Antisense treatment did not ameliorate proteinuria or pathologic manifestations of diabetic nephropathy, including podocyte loss. These studies indicate that pathologic manifestations of diabetic nephropathy are unlikely to be reduced by lipid-lowering therapeutics alone, but does not preclude a role for such interventions to be used in conjunction with other therapeutics commonly employed in the treatment of diabetes and its complications.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/metabolismo , Hipertrigliceridemia/metabolismo , Animales , Apolipoproteína C-III/genética , Apolipoproteína C-III/metabolismo , Diabetes Mellitus Experimental/metabolismo , Femenino , Leptina/genética , Masculino , Ratones , Ratones Obesos , Oligonucleótidos Antisentido , Podocitos/metabolismo , Podocitos/patología
5.
Nutr Metab Cardiovasc Dis ; 31(8): 2253-2272, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34059383

RESUMEN

AIM: Diabetic nephropathy (DN) is one of the worst complications of diabetes. Despite a growing number of DN metabolite profiling studies, most studies are suffering from inconsistency in their findings. The main goal of this meta-analysis was to reach to a consensus panel of significantly dysregulated metabolites as potential biomarkers in DN. DATA SYNTHESIS: To identify the significant dysregulated metabolites, meta-analysis was performed by "vote-counting rank" and "robust rank aggregation" strategies. Bioinformatics analyses were performed to identify the most affected genes and pathways. Among 44 selected studies consisting of 98 metabolite profiles, 17 metabolites (9 up-regulated and 8 down-regulated metabolites), were identified as significant ones by both the meta-analysis strategies (p-value<0.05 and OR>2 or <0.5) and selected as DN metabolite meta-signature. Furthermore, enrichment analyses confirmed the involvement of various effective biological pathways in DN pathogenesis, such as urea cycle, TCA cycle, glycolysis, and amino acid metabolisms. Finally, by performing a meta-analysis over existing time-course studies in DN, the results indicated that lactic acid, hippuric acid, allantoin (in urine), and glutamine (in blood), are the topmost non-invasive early diagnostic biomarkers. CONCLUSION: The identified metabolites are potentially involved in diabetic nephropathy pathogenesis and could be considered as biomarkers or drug targets in the disease. PROSPERO REGISTRATION NUMBER: CRD42020197697.


Asunto(s)
Nefropatías Diabéticas/diagnóstico , Metaboloma , Metabolómica , Animales , Biomarcadores/sangre , Biomarcadores/orina , Nefropatías Diabéticas/sangre , Nefropatías Diabéticas/orina , Diagnóstico Precoz , Humanos , Valor Predictivo de las Pruebas
6.
Am J Physiol Renal Physiol ; 318(5): F1295-F1305, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32249614

RESUMEN

Podocyte loss and proteinuria are both key features of human diabetic nephropathy (DN). The leptin-deficient BTBR mouse strain with the ob/ob mutation develops progressive weight gain, type 2 diabetes, and diabetic nephropathy that has many features of advanced human DN, including increased mesangial matrix, mesangiolysis, podocyte loss, and proteinuria. Selective antagonism of the endothelin-1 type A receptor (ETAR) by atrasentan treatment in combination with renin-angiotensin-aldosterone system inhibition with losartan has been shown to have the therapeutic benefit of lowering proteinuria in patients with DN, but the underlying mechanism for this benefit is not well understood. Using a similar therapeutic approach in diabetic BTBR ob/ob mice, this treatment regimen significantly increased glomerular podocyte number compared with diabetic BTBR ob/ob controls and suggested that parietal epithelial cells were a source for podocyte restoration. Atrasentan treatment alone also increased podocyte number but to a lesser degree. Mice treated with atrasentan demonstrated a reduction in proteinuria, matching the functional improvement reported in humans. This is a first demonstration that treatment with the highly selective ETAR antagonist atrasentan can lead to restoration of the diminished podocyte number characteristic of DN in humans and thereby underlies the reduction in proteinuria in patients with diabetes undergoing similar treatment. The benefit of ETAR antagonism in DN extended to a decrease in mesangial matrix as measured by a reduction in accumulations of collagen type IV in both the atrasentan and atrasentan + losartan-treated groups compared with untreated controls.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Atrasentán/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Nefropatías Diabéticas/prevención & control , Antagonistas de los Receptores de la Endotelina A/farmacología , Losartán/farmacología , Podocitos/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Ratones , Fosforilación , Podocitos/metabolismo , Podocitos/patología , Proteinuria/metabolismo , Proteinuria/patología , Proteinuria/prevención & control , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
J Am Soc Nephrol ; 30(3): 421-441, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30760496

RESUMEN

BACKGROUND: Linking genetic risk loci identified by genome-wide association studies (GWAS) to their causal genes remains a major challenge. Disease-associated genetic variants are concentrated in regions containing regulatory DNA elements, such as promoters and enhancers. Although researchers have previously published DNA maps of these regulatory regions for kidney tubule cells and glomerular endothelial cells, maps for podocytes and mesangial cells have not been available. METHODS: We generated regulatory DNA maps (DNase-seq) and paired gene expression profiles (RNA-seq) from primary outgrowth cultures of human glomeruli that were composed mainly of podocytes and mesangial cells. We generated similar datasets from renal cortex cultures, to compare with those of the glomerular cultures. Because regulatory DNA elements can act on target genes across large genomic distances, we also generated a chromatin conformation map from freshly isolated human glomeruli. RESULTS: We identified thousands of unique regulatory DNA elements, many located close to transcription factor genes, which the glomerular and cortex samples expressed at different levels. We found that genetic variants associated with kidney diseases (GWAS) and kidney expression quantitative trait loci were enriched in regulatory DNA regions. By combining GWAS, epigenomic, and chromatin conformation data, we functionally annotated 46 kidney disease genes. CONCLUSIONS: We demonstrate a powerful approach to functionally connect kidney disease-/trait-associated loci to their target genes by leveraging unique regulatory DNA maps and integrated epigenomic and genetic analysis. This process can be applied to other kidney cell types and will enhance our understanding of genome regulation and its effects on gene expression in kidney disease.

8.
Am J Physiol Renal Physiol ; 316(6): F1201-F1210, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30995111

RESUMEN

Renal Na+-glucose cotransporter SGLT1 mediates glucose reabsorption in the late proximal tubule, a hypoxia-sensitive tubular segment that enters the outer medulla. Gene deletion in mice (Sglt1-/-) was used to determine the role of the cotransporter in acute kidney injury induced by ischemia-reperfusion (IR), including the initial injury and subsequent recovery phase. On days 1 and 16 after IR, absolute and fractional urinary glucose excretion remained greater in Sglt1-/- mice versus wild-type (WT) littermates, consistent with a sustained contribution of SGLT1 to tubular glucose reabsorption in WT mice. Absence of SGLT1 did not affect the initial kidney impairment versus WT mice, as indicated by similar increases on day 1 in plasma concentrations of creatinine and urinary excretion of the tubular injury marker kidney injury molecule-1 as well as a similar rise in plasma osmolality and fall in urine osmolality as indicators of impaired urine concentration. Recovery of kidney function on days 14/16, however, was improved in Sglt1-/- versus WT mice, as indicated by lower plasma creatinine, higher glomerula filtration rate (by FITC-sinistrin in awake mice), and more completely restored urine and plasma osmolality. This was associated with a reduced tubular injury score in the cortex and outer medulla, better preserved renal mRNA expression of tubular transporters (Sglt2 and Na+-K+-2Cl- cotransporter Nkcc2), and a lesser rise in renal mRNA expression of markers of injury, inflammation, and fibrosis [kidney injury molecule-1, chemokine (C-C motif) ligand 2, fibronectin 1, and collagen type I-α1] in Sglt1-/- versus WT mice. These results suggest that SGLT1 activity in the late proximal tubule may have deleterious effects during recovery of IR-induced acute kidney injury and identify SGLT1 as a potential therapeutic target.


Asunto(s)
Lesión Renal Aguda/metabolismo , Tasa de Filtración Glomerular , Glucosa/metabolismo , Túbulos Renales Proximales/metabolismo , Reabsorción Renal , Daño por Reperfusión/metabolismo , Transportador 1 de Sodio-Glucosa/deficiencia , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Recuperación de la Función , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transportador 1 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/genética , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Factores de Tiempo
9.
Am J Pathol ; 188(2): 343-352, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29154962

RESUMEN

Diabetic kidney disease and atherosclerotic disease are major causes of morbidity and mortality associated with type 2 diabetes (T2D), and diabetic kidney disease is a major cardiovascular risk factor. The black and tan, brachyury (BTBR) mouse strain with leptin deficiency (Lepob) has emerged as one of the best models of human diabetic kidney disease. However, no T2D mouse model of combined diabetic kidney disease and atherosclerosis exists. Our goal was to generate such a model. To this end, the low-density lipoprotein (LDL) receptor was targeted for degradation via inducible degrader of the LDL receptor (IDOL) overexpression, using liver-targeted adenoassociated virus serotype DJ/8 (AAV-DJ/8) in BTBR wild-type and BTBR Lepob mice. Liver-targeted IDOL-AAV-DJ/8 increased plasma LDL cholesterol compared with the control enhanced green fluorescent protein AAV-DJ/8. IDOL-induced dyslipidemia caused formation of atherosclerotic lesions of an intermediate stage, which contained both macrophages and smooth muscle cells. BTBR Lepob mice exhibited diabetic kidney disease. IDOL-induced dyslipidemia worsened albuminuria and glomerular macrophage accumulation but had no effect on mesangial expansion or podocyte numbers. Thus, by inducing hepatic degradation of the LDL receptor, we generated a T2D model of combined kidney disease and atherosclerosis. This model provides a new tool to study mechanisms, interactions, and treatment strategies of kidney disease and atherosclerosis in T2D.


Asunto(s)
Aterosclerosis/etiología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/etiología , Animales , Aterosclerosis/sangre , Aterosclerosis/patología , Colesterol/sangre , Dependovirus/genética , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 2/sangre , Nefropatías Diabéticas/sangre , Modelos Animales de Enfermedad , Dislipidemias/sangre , Dislipidemias/complicaciones , Vectores Genéticos , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Receptores de LDL/biosíntesis , Receptores de LDL/deficiencia , Receptores de LDL/genética
10.
Kidney Int ; 94(4): 728-740, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29907458

RESUMEN

B cells are known to promote the pathogenesis of systemic lupus erythematosus (SLE) via the production of pathogenic anti-nuclear antibodies. However, the signals required for autoreactive B cell activation and the immune mechanisms whereby B cells impact lupus nephritis pathology remain poorly understood. The B cell survival cytokine B cell activating factor of the TNF Family (BAFF) has been implicated in the pathogenesis of SLE and lupus nephritis in both animal models and human clinical studies. Although the BAFF receptor has been predicted to be the primary BAFF family receptor responsible for BAFF-driven humoral autoimmunity, in the current study we identify a critical role for signals downstream of Transmembrane Activator and CAML Interactor (TACI) in BAFF-dependent lupus nephritis. Whereas transgenic mice overexpressing BAFF develop progressive membranoproliferative glomerulonephritis, albuminuria and renal dysfunction, TACI deletion in BAFF-transgenic mice provided long-term (about 1 year) protection from renal disease. Surprisingly, disease protection in this context was not explained by complete loss of glomerular immune complex deposits. Rather, TACI deletion specifically reduced endocapillary, but not mesangial, immune deposits. Notably, although excess BAFF promoted widespread breaks in B cell tolerance, BAFF-transgenic antibodies were enriched for RNA- relative to DNA-associated autoantigen reactivity. These RNA-associated autoantibody specificities were specifically reduced by TACI or Toll-like receptor 7 deletion. Thus, our study provides important insights into the autoantibody specificities driving proliferative lupus nephritis, and suggests that TACI inhibition may be novel and effective treatment strategy in lupus nephritis.


Asunto(s)
Autoanticuerpos/sangre , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Nefritis Lúpica/genética , Ribonucleoproteínas/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Albuminuria/genética , Albuminuria/orina , Animales , Factor Activador de Células B/sangre , Factor Activador de Células B/inmunología , Linfocitos B/inmunología , Creatinina/orina , Progresión de la Enfermedad , Femenino , Hipergammaglobulinemia/genética , Inmunoglobulinas/sangre , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Activación de Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
11.
J Am Soc Nephrol ; 28(10): 3076-3088, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28724774

RESUMEN

Stenosis from venous neointimal hyperplasia is common in native arteriovenous fistulas (AVFs). However, the preexisting histologic characteristics of veins at fistula creation, and associations thereof with baseline patient factors, have not been well characterized. In this study, we conducted histologic analysis of a segment of the vein used for anastomosis creation, obtained during AVF creation from 554 of the 602 participants in the multicenter Hemodialysis Fistula Maturation Cohort Study. We quantified intimal and medial areas and lengths of the internal and external elastic lamina by morphometry and assessed venous wall cells by immunohistochemistry, extracellular matrix with Movat stain, and calcium deposition by alizarin red stain. We also studied a representative subset of veins for markers of monocyte/macrophage content, cell proliferation, apoptosis, and neoangiogenesis. Neointima occupied >20% of the lumen in 57% of fully circumferential vein samples, and neointimal hyperplasia associated positively with age and inversely with black race. The neointima was usually irregularly thickened, sometimes concentric, and contained α-smooth muscle actin-expressing cells of smooth muscle or myofibroblast origin. Proteoglycans admixed with lesser amounts of collagen constituted the predominant matrix in the neointima. In 82% of vein samples, the media of vessel walls contained large aggregates of collagen. A minority of veins expressed markers of inflammation, cell proliferation, cell death, calcification, or neoangiogenesis. In conclusion, we observed preexisting abnormalities, including neointimal hyperplasia and prominent accumulation of extracellular matrix, in veins used for AVF creation from a substantial proportion of this cohort.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Neointima/patología , Calcificación Vascular/patología , Venas/patología , Adulto , Anciano , Femenino , Humanos , Fallo Renal Crónico/patología , Fallo Renal Crónico/terapia , Masculino , Persona de Mediana Edad , Diálisis Renal
12.
Kidney Int ; 91(5): 1126-1145, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28063595

RESUMEN

Although age-associated changes in kidney glomerular architecture have been described in mice and man, the mechanisms are unknown. It is unclear if these changes can be prevented or even reversed by systemic therapies administered at advanced age. Using light microscopy and transmission electron microscopy, our results showed glomerulosclerosis with injury to mitochondria in glomerular epithelial cells in mice aged 26 months (equivalent to a 79-year-old human). To test the hypothesis that reducing mitochondrial damage in late age would result in lowered glomerulosclerosis, we administered the mitochondrial targeted peptide, SS-31, to aged mice. Baseline (24-month-old) mice were randomized to receive 8 weeks of SS-31, or saline, and killed at 26 months of age. SS-31 treatment improved age-related mitochondrial morphology and glomerulosclerosis. Assessment of glomeruli revealed that SS-31 reduced senescence (p16, senescence-associated-ß-Gal) and increased the density of parietal epithelial cells. However, SS-31 treatment reduced markers of parietal epithelial cell activation (Collagen IV, pERK1/2, and α-smooth muscle actin). SS-31 did not impact podocyte density, but it reduced markers of podocyte injury (desmin) and improved cytoskeletal integrity (synaptopodin). This was accompanied by higher glomerular endothelial cell density (CD31). Thus, despite initiating therapy in late-age mice, a short course of SS-31 has protective benefits on glomerular mitochondria, accompanied by temporal changes to the glomerular architecture. This systemic pharmacological intervention in old-aged animals limits glomerulosclerosis and senescence, reduces parietal epithelial cell activation, and improves podocyte and endothelial cell integrity.


Asunto(s)
Envejecimiento/efectos de los fármacos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Mitocondrias/efectos de los fármacos , Oligopéptidos/farmacología , Actinas/metabolismo , Envejecimiento/fisiología , Animales , Colágeno Tipo IV/metabolismo , Desmina/metabolismo , Células Endoteliales/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Glomérulos Renales/citología , Masculino , Ratones , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica de Transmisión , Mitocondrias/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Podocitos/efectos de los fármacos , Esclerosis
13.
J Immunol ; 192(10): 4525-32, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24711620

RESUMEN

Systemic lupus erythematosus is a multisystem autoimmune disease characterized by autoantibodies targeting nucleic acid-associated Ags. The endosomal TLRs TLR7 and TLR9 are critical for generation of Abs targeting RNA- or DNA-associated Ags, respectively. In murine lupus models, deletion of TLR7 limits autoimmune inflammation, whereas deletion of TLR9 exacerbates disease. Whether B cell or myeloid TLR7/TLR9 signaling is responsible for these effects has not been fully addressed. In this study, we use a chimeric strategy to evaluate the effect of B cell-intrinsic deletion of TLR7 versus TLR9 in parallel lupus models. We demonstrate that B cell-intrinsic TLR7 deletion prevents RNA-associated Ab formation, decreases production of class-switched Abs targeting nonnuclear Ags, and limits systemic autoimmunity. In contrast, B cell-intrinsic TLR9 deletion results in decreased DNA-reactive Ab, but increased Abs targeting a broad range of systemic autoantigens. Further, we demonstrate that B cell-intrinsic TLR9 deletion results in increased systemic inflammation and immune complex glomerulonephritis, despite intact TLR signaling within the myeloid compartment. These data stress the critical importance of dysregulated B cell-intrinsic TLR signaling in the pathogenesis of systemic lupus erythematosus.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Glicoproteínas de Membrana/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Animales , Anticuerpos Antinucleares/genética , Linfocitos B/patología , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , ARN/genética , ARN/inmunología , Transducción de Señal/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética
14.
Lab Invest ; 95(3): 250-62, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25531567

RESUMEN

Inflammatory pathways are central mechanisms in diabetic kidney disease (DKD). Serum amyloid A (SAA) is increased by chronic inflammation, but SAA has not been previously evaluated as a potential DKD mediator. The aims of this study were to determine whether SAA is increased in human DKD and corresponding mouse models and to assess effects of SAA on podocyte inflammatory responses. SAA was increased in the plasma of people with DKD characterized by overt proteinuria and inversely correlated with estimated glomerular filtration rate (creatinine-based CKD-EPI). SAA was also elevated in plasma of diabetic mouse models including type 1 diabetes (streptozotocin/C57BL/6) and type 2 diabetes (BTBR-ob/ob). SAA mRNA (Nephromine) was increased in human DKD compared with non-diabetic and/or glomerular disease controls (glomerular fold change 1.5, P=0.017; tubulointerstitium fold change 1.4, P=0.021). The kidneys of both diabetic mouse models also demonstrated increased SAA mRNA (quantitative real-time PCR) expression compared with non-diabetic controls (type 1 diabetes fold change 2.9; type 2 diabetes fold change 42.5, P=0.009; interaction by model P=0.57). Humans with DKD and the diabetic mouse models exhibited extensive SAA protein deposition in the glomeruli and tubulointerstitium in similar patterns by immunohistochemistry. SAA localized within podocytes of diabetic mice. Podocytes exposed to advanced glycation end products, metabolic mediators of inflammation in diabetes, increased expression of SAA mRNA (fold change 15.3, P=0.004) and protein (fold change 38.4, P=0.014). Podocytes exposed to exogenous SAA increased NF-κB activity, and pathway array analysis revealed upregulation of mRNA for NF-κB-dependent targets comprising numerous inflammatory mediators, including SAA itself (fold change 17.0, P=0.006). Inhibition of NF-κB reduced these pro-inflammatory responses. In conclusion, SAA is increased in the blood and produced in the kidneys of people with DKD and corresponding diabetic mouse models. Podocytes are likely to be key responder cells to SAA-induced inflammation in the diabetic kidney. SAA is a compelling candidate for DKD therapeutic and biomarker discovery.


Asunto(s)
Nefropatías Diabéticas/sangre , Inflamación/sangre , Podocitos/metabolismo , Proteína Amiloide A Sérica/metabolismo , Animales , Antioxidantes/farmacología , Células Cultivadas , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 2/sangre , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/sangre , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Podocitos/efectos de los fármacos , Pirrolidinas/farmacología , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/farmacología , Tiocarbamatos/farmacología
15.
Kidney Int ; 88(5): 1099-107, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26376129

RESUMEN

Reversal of diabetic nephropathy (DN) has been achieved in humans and mice, but only rarely and under special circumstances. As progression of DN is related to podocyte loss, reversal of DN requires restoration of podocytes. Here, we identified and quantified potential glomerular progenitor cells that could be a source for restored podocytes. DN was identified in 31 human renal biopsy cases and separated into morphologically early or advanced lesions. Markers of podocytes (WT-1, p57), parietal epithelial cells (PECs) (claudin-1), and cell proliferation (Ki-67) were identified by immunohistochemistry. Podocyte density was progressively reduced with DN. Cells marking as podocytes (p57) were present infrequently on Bowman's capsule in controls, but significantly increased in histologically early DN. Ki-67-expressing cells were identified on the glomerular tuft and Bowman's capsule in DN, but rarely in controls. Cells marking as PECs were present on the glomerular tuft, particularly in morphologically advanced DN. These findings show evidence of phenotypic plasticity in podocyte and PEC populations and are consistent with studies in the BTBR ob/ob murine model in which reversibility of DN occurs with podocytes potentially regenerating from PEC precursors. Thus, our findings support, but do not prove, that podocytes may regenerate from PEC progenitors in human DN. If so, progression of DN may represent a modifiable net balance between podocyte loss and regeneration.


Asunto(s)
Plasticidad de la Célula , Nefropatías Diabéticas/patología , Podocitos/citología , Regeneración , Células Madre/fisiología , Cápsula Glomerular/química , Cápsula Glomerular/patología , Proliferación Celular , Claudina-1/análisis , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/análisis , Células Epiteliales/química , Células Epiteliales/citología , Células Epiteliales/fisiología , Humanos , Antígeno Ki-67/análisis , Glomérulos Renales/patología , Proteínas de Microfilamentos/análisis , Fenotipo , Podocitos/química , Podocitos/fisiología , Estudios Retrospectivos , Células Madre/citología , Proteínas WT1/análisis
16.
Curr Opin Nephrol Hypertens ; 24(3): 245-51, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25887903

RESUMEN

PURPOSE OF REVIEW: Foam cells in human glomeruli can be encountered in various renal diseases including focal segmental glomerulosclerosis and diabetic nephropathy. Although foam cells are key participants in atherosclerosis, surprisingly little is known about their pathogenicity in the kidney. We review our understanding (or lack thereof) of foam cells in the kidney, as well as insights gained in studies of foam cells and macrophages involved in atherosclerosis to suggest areas of investigation that will allow better characterization of the role of these cells in renal disease. RECENT FINDINGS: There is a general dearth of animal models of disease with renal foam cell accumulation, limiting progress in our understanding of the pathobiology of these cells. Recent genetic modifications of hyperlipidemic mice have resulted in some new disease models with renal foam cell accumulation. Recent studies have challenged older paradigms by findings that indicate that many tissue macrophages are derived from cells permanently residing in the tissue from birth rather than circulating monocytes. SUMMARY: Renal foam cells remain an enigma. Extrapolating from studies of atherosclerosis suggests that therapeutics targeting mitochondrial reactive oxygen species production, or modulating cholesterol and lipoprotein uptake or egress from these cells, may prove beneficial for kidney diseases in which foam cells are present.


Asunto(s)
Aterosclerosis/patología , Células Espumosas/citología , Enfermedades Renales/patología , Glomérulos Renales/citología , Macrófagos/citología , Monocitos/citología , Animales , Aterosclerosis/complicaciones , Humanos
17.
J Immunol ; 190(6): 2536-43, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23382559

RESUMEN

TLR7 activation is implicated in the pathogenesis of systemic lupus erythematosus. Mice that overexpress TLR7 develop a lupus-like disease with autoantibodies and glomerulonephritis and early death. To determine whether degradation of the TLR7 ligand RNA would alter the course of disease, we created RNase A transgenic (Tg) mice. We then crossed the RNase Tg to TLR7 Tg mice to create TLR7 × RNase double Tg (DTg) mice. DTg mice had a significantly increased survival associated with reduced activation of T and B lymphocytes and reduced kidney deposition of IgG and C3. We observed massive hepatic inflammation and cell death in TLR7 Tg mice. In contrast, hepatic inflammation and necrosis were strikingly reduced in DTg mice. These findings indicate that high concentrations of serum RNase protect against immune activation and inflammation associated with TLR7 stimulation and that RNase may be a useful therapeutic strategy in the prevention or treatment of inflammation in systemic lupus erythematosus and, possibly, liver diseases.


Asunto(s)
Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ribonucleasa Pancreática/genética , Receptor Toll-Like 7/biosíntesis , Receptor Toll-Like 7/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Animales , Bovinos , Células Cultivadas , Células Madre Embrionarias , Hepatitis/enzimología , Hepatitis/inmunología , Hepatitis/patología , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/prevención & control , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/mortalidad , Lupus Eritematoso Sistémico/prevención & control , Masculino , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Ribonucleasa Pancreática/sangre , Ribonucleasa Pancreática/fisiología , Bazo/enzimología , Bazo/inmunología , Bazo/patología , Análisis de Supervivencia , Receptor Toll-Like 7/fisiología
18.
Lab Invest ; 94(8): 851-62, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24955894

RESUMEN

Piperidine-based peroxisome proliferator-activated receptor-α agonists are agents that are efficacious in improving lipid, glycemic, and inflammatory indicators in diabetes and obesity. This study sought to determine whether CP-900691 ((S)-3-[3-(1-carboxy-1-methyl-ethoxy)-phenyl]-piperidine-1-carboxylic acid 4-trifluoromethyl-benzyl ester; CP), a member of this novel class of agents, by decreasing plasma triglycerides, could prevent diabetic nephropathy in the Black and Tan, BRachyuric (BTBR) ob/ob mouse model of type 2 diabetes mellitus. Four-week old female BTBR WT and BTBR ob/ob mice received either regular chow or one containing CP (3 mg/kg per day) for 14 weeks. CP elevated plasma high-density lipoprotein, albuminuria, and urinary excretion of 8-epi PGF(2α), a product of the nonenzymatic metabolism of arachidonic acid and whose production is elevated in oxidative stress, in BTBR WT mice. In BTBR ob/ob mice, CP reduced plasma triglycerides and non-esterified fatty acids, fasting blood glucose, body weight, and plasma interleukin-6, while concomitantly improving insulin resistance. Despite these beneficial metabolic effects, CP had no effect on elevated plasma insulin, 8-epi PGF(2α) excretion, and albuminuria, and surprisingly, did not ameliorate the development of diabetic nephropathy, having no effect on the accumulation of renal macrophages, glomerular hypertrophy, and increased mesangial matrix expansion. In addition, CP did not increase plasma high-density lipoprotein in BTBR ob/ob mice, while paradoxically increasing total cholesterol levels. These findings indicate that 8-epi PGF(2α), possibly along with hyperinsulinemia and inflammatory and dysfunctional lipoproteins, is integral to the development of diabetic nephropathy and should be considered as a potential target of therapy in the treatment of diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Nefropatías Diabéticas/prevención & control , Hipoglucemiantes/uso terapéutico , Riñón/efectos de los fármacos , Obesidad/prevención & control , PPAR alfa/agonistas , Piperidinas/uso terapéutico , Propionatos/uso terapéutico , Albuminuria/complicaciones , Albuminuria/fisiopatología , Albuminuria/prevención & control , Animales , Fármacos Antiobesidad/efectos adversos , Fármacos Antiobesidad/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/fisiopatología , Dinoprost/agonistas , Dinoprost/análogos & derivados , Dinoprost/orina , Progresión de la Enfermedad , Femenino , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/inmunología , Mesangio Glomerular/metabolismo , Mesangio Glomerular/patología , Hipercolesterolemia/inducido químicamente , Hipercolesterolemia/complicaciones , Hipercolesterolemia/fisiopatología , Hipercolesterolemia/prevención & control , Hipertrigliceridemia/complicaciones , Hipertrigliceridemia/prevención & control , Hipertrofia , Hipoglucemiantes/efectos adversos , Hipolipemiantes/efectos adversos , Hipolipemiantes/uso terapéutico , Resistencia a la Insulina , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos , Ratones Obesos , Obesidad/complicaciones , PPAR alfa/metabolismo , Piperidinas/efectos adversos , Propionatos/efectos adversos
19.
Int J Cancer ; 134(4): 778-88, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23929039

RESUMEN

Cirrhosis is the primary risk factor for the development of hepatocellular carcinoma (HCC), yet the mechanisms by which cirrhosis predisposes to carcinogenesis are poorly understood. Using a mouse model that recapitulates many aspects of the pathophysiology of human liver disease, we explored the mechanisms by which changes in the liver microenvironment induce dysplasia and HCC. Hepatic expression of platelet-derived growth factor C (PDGF-C) induces progressive fibrosis, chronic inflammation, neoangiogenesis and sinusoidal congestion, as well as global changes in gene expression. Using reporter mice, immunofluorescence, immunohistochemistry and liver cell isolation, we demonstrate that receptors for PDGF-CC are localized on hepatic stellate cells (HSCs), which proliferate, and transform into myofibroblast-like cells that deposit extracellular matrix and lead to production of growth factors and cytokines. We demonstrate induction of cytokine genes at 2 months, and stromal cell-derived hepatocyte growth factors that coincide with the onset of dysplasia at 4 months. Our results support a paracrine signaling model wherein hepatocyte-derived PDGF-C stimulates widespread HSC activation throughout the liver leading to chronic inflammation, liver injury and architectural changes. These complex changes to the liver microenvironment precede the development of HCC. Further, increased PDGF-CC levels were observed in livers of patients with nonalcoholic fatty steatohepatitis and correlate with the stage of disease, suggesting a role for this growth factor in chronic liver disease in humans. PDGF-C transgenic mice provide a unique model for the in vivo study of tumor-stromal interactions in the liver.


Asunto(s)
Carcinoma Hepatocelular/patología , Hígado Graso/patología , Células Estrelladas Hepáticas/patología , Neoplasias Hepáticas/patología , Linfocinas/metabolismo , Comunicación Paracrina , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células del Estroma/patología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Estudios de Cohortes , Citocinas/genética , Citocinas/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Técnicas para Inmunoenzimas , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Linfocinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Derivado de Plaquetas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/metabolismo
20.
Am J Respir Crit Care Med ; 188(1): 42-50, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23614662

RESUMEN

RATIONALE: Indirect airway hyperresponsiveness (AHR) is a fundamental feature of asthma that is manifest as exercise-induced bronchoconstriction (EIB). Secreted phospholipase A2 group X (sPLA2-X) plays a key role in regulating eicosanoid formation and the development of inflammation and AHR in murine models. OBJECTIVES: We sought to examine sPLA2-X in the airway epithelium and airway wall of patients with asthma, the relationship to AHR in humans, and the regulation and function of sPLA2-X within the epithelium. METHODS: We precisely phenotyped 34 patients with asthma (19 with and 15 without EIB) and 10 normal control subjects to examine in vivo differences in epithelial gene expression, quantitative morphometry of endobronchial biopsies, and levels of secreted protein. The regulation of sPLA2-X gene (PLA2G10) expression was examined in primary airway epithelial cell cultures. The function of epithelial sPLA2-X in eicosanoid formation was examined using PLA2 inhibitors and murine tracheal epithelial cells with Pla2g10 deletion. MEASUREMENTS AND MAIN RESULTS: We found that sPLA2-X protein is increased in the airways of patients with asthma and that epithelial-derived sPLA2-X may be increased in association with indirect AHR. The expression of sPLA2-X increases during in vitro epithelial differentiation; is regulated by inflammatory signals including tumor necrosis factor, IL-13, and IL-17; and is both secreted from the epithelium and directly participates in the release of arachidonic acid by epithelial cells. CONCLUSIONS: These data reveal a relationship between epithelial-derived sPLA2-X and indirect AHR in asthma and that sPLA2-X serves as an epithelial regulator of inflammatory eicosanoid formation. Therapies targeting epithelial sPLA2-X may be useful in asthma.


Asunto(s)
Asma/genética , Asma/inmunología , Células Epiteliales/inmunología , Fosfolipasas A2 Grupo X/genética , Fosfolipasas A2 Grupo X/inmunología , Adolescente , Adulto , Animales , Asma Inducida por Ejercicio/genética , Asma Inducida por Ejercicio/inmunología , Hiperreactividad Bronquial/genética , Hiperreactividad Bronquial/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA