Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Med ; 29(1): 92, 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37415117

RESUMEN

BACKGROUND: Diabetic nephropathy (DN) is a major complication of diabetes mellitus. Clinical reports indicate that smoking is a significant risk factor for chronic kidney disease, and the tobacco epidemic exacerbates kidney damage in patients with DN. However, the underlying molecular mechanisms remain unclear. METHOD: In the present study, we used a diabetic mouse model to investigate the molecular mechanisms for nicotine-exacerbated DN. Twelve-week-old female mice were injected with streptozotocin (STZ) to establish a hyperglycemic diabetic model. After four months, the control and hyperglycemic diabetic mice were further divided into four groups (control, nicotine, diabetic mellitus, nicotine + diabetic mellitus) by intraperitoneal injection of nicotine or PBS. After two months, urine and blood were collected for kidney injury assay, and renal tissues were harvested for further molecular assays using RNA-seq analysis, real-time PCR, Western blot, and immunohistochemistry. In vitro studies, we used siRNA to suppress Grem1 expression in human podocytes. Then we treated them with nicotine and high glucose to compare podocyte injury. RESULT: Nicotine administration alone did not cause apparent kidney injury, but it significantly increased hyperglycemia-induced albuminuria, BUN, plasma creatinine, and the kidney tissue mRNA expression of KIM-1 and NGAL. Results from RNA-seq analysis, real-time PCR, Western blot, and immunohistochemistry analysis revealed that, compared to hyperglycemia or nicotine alone, the combination of nicotine treatment and hyperglycemia significantly increased the expression of Grem1 and worsened DN. In vitro experiments, suppression of Grem1 expression attenuated nicotine-exacerbated podocyte injury. CONCLUSION: Grem1 plays a vital role in nicotine-exacerbated DN. Grem1 may be a potential therapeutic target for chronic smokers with DN.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Hiperglucemia , Humanos , Ratones , Femenino , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/inducido químicamente , Regulación hacia Arriba , Nicotina/efectos adversos , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inducido químicamente , Hiperglucemia/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo
2.
Platelets ; 31(3): 399-402, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31146647

RESUMEN

Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder in which autoantibodies and/or autoreactive T cells destroy platelets and megakaryocytes in the spleen and bone marrow, respectively. Thrombopoietin receptor agonists (TPO-RA e.g. Romiplostim and Eltrombopag) have made a substantial contribution to the treatment of patients with ITP, which are refractory to first-line treatments and approximately 30% demonstrate sustained elevated platelet counts after drug tapering. How TPO-RA induce these sustained responses is not known. We analyzed the efficacy of a murine TPO-RA in a well-established murine model of active ITP. Treatment with TPO-RA (10 ug/kg, based on pilot dose escalation experiments) significantly raised the platelet counts in ITP-mice. Immunomodulation was assessed by measuring serum IgG anti-platelet antibody levels; TPO-RA-treated mice had significantly reduced IgG anti-platelet antibodies despite the increasing platelet counts. These results suggest that TPO-RA is not only an efficacious therapy but also reduces anti-platelet humoral immunity in ITP.


Asunto(s)
Autoanticuerpos/inmunología , Plaquetas/inmunología , Plaquetas/metabolismo , Recuento de Plaquetas , Púrpura Trombocitopénica Idiopática/etiología , Púrpura Trombocitopénica Idiopática/metabolismo , Receptores de Trombopoyetina/agonistas , Animales , Autoinmunidad , Biopsia , Plaquetas/efectos de los fármacos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Inmunomodulación , Ratones , Ratones Noqueados , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/patología
3.
Am J Pathol ; 188(11): 2508-2528, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30201495

RESUMEN

Human parietal epithelial cells (PECs) are progenitor cells that sustain podocyte homeostasis. We hypothesized that the lack of apolipoprotein (APO) L1 ensures the PEC phenotype, but its induction initiates PEC transition (expression of podocyte markers). APOL1 expression and down-regulation of miR193a coincided with the expression of podocyte markers during the transition. The induction of APOL1 also stimulated transition markers in human embryonic kidney cells (cells with undetectable APOL1 protein expression). APOL1 silencing in PECs up-regulated miR193a expression, suggesting the possibility of a reciprocal feedback relationship between APOL1 and miR193a. HIV, interferon-γ, and vitamin D receptor agonist down-regulated miR193a expression and induced APOL1 expression along with transition markers in PECs. Luciferase assay suggested a putative interaction between miR193a and APOL1. Since silencing of APOL1 attenuated HIV-, vitamin D receptor agonist-, miR193a inhibitor-, and interferon-γ-induced expression of transition markers, APOL1 appears to be a critical functional constituent of the miR193a- APOL1 axis in PECs. This notion was confirmed by further enhanced expression of PEC markers in APOL1 mRNA-silenced PECs. In vivo studies, glomeruli in patients with HIV, and HIV/APOL1 transgenic mice had foci of PECs expressing synaptopodin, a transition marker. APOL1 likely regulates PEC molecular phenotype through modulation of miR193a expression, and APOL1 and miR193a share a reciprocal feedback relationship.


Asunto(s)
Nefropatía Asociada a SIDA/patología , Apolipoproteína L1/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Glomérulos Renales/patología , MicroARNs/genética , Nefropatía Asociada a SIDA/metabolismo , Nefropatía Asociada a SIDA/virología , Animales , Apolipoproteína L1/genética , Estudios de Casos y Controles , Células Epiteliales/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Glomérulos Renales/metabolismo , Ratones , Ratones Transgénicos
4.
Blood ; 129(18): 2557-2569, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28202460

RESUMEN

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related fatalities and is characterized by acute respiratory distress following blood transfusion. Donor antibodies are frequently involved; however, the pathogenesis and protective mechanisms in the recipient are poorly understood, and specific therapies are lacking. Using newly developed murine TRALI models based on injection of anti-major histocompatibility complex class I antibodies, we found CD4+CD25+FoxP3+ T regulatory cells (Tregs) and CD11c+ dendritic cells (DCs) to be critical effectors that protect against TRALI. Treg or DC depletion in vivo resulted in aggravated antibody-mediated acute lung injury within 90 minutes with 60% mortality upon DC depletion. In addition, resistance to antibody-mediated TRALI was associated with increased interleukin-10 (IL-10) levels, and IL-10 levels were found to be decreased in mice suffering from TRALI. Importantly, IL-10 injection completely prevented and rescued the development of TRALI in mice and may prove to be a promising new therapeutic approach for alleviating lung injury in this serious complication of transfusion.


Asunto(s)
Lesión Pulmonar Aguda , Células Dendríticas/inmunología , Interleucina-10 , Linfocitos T Reguladores/inmunología , Reacción a la Transfusión , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/prevención & control , Animales , Anticuerpos/inmunología , Células Dendríticas/patología , Interleucina-10/inmunología , Interleucina-10/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Linfocitos T Reguladores/patología
5.
Am J Physiol Renal Physiol ; 314(5): F832-F843, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357419

RESUMEN

The loss of podocyte (PD) molecular phenotype is an important feature of diabetic podocytopathy. We hypothesized that high glucose (HG) induces dedifferentiation in differentiated podocytes (DPDs) through alterations in the apolipoprotein (APO) L1-microRNA (miR) 193a axis. HG-induced DPD dedifferentiation manifested in the form of downregulation of Wilms' tumor 1 (WT1) and upregulation of paired box 2 (PAX2) expression. WT1-silenced DPDs displayed enhanced expression of PAX2. Immunoprecipitation of DPD cellular lysates with anti-WT1 antibody revealed formation of WT1 repressor complexes containing Polycomb group proteins, enhancer of zeste homolog 2, menin, and DNA methyltransferase (DNMT1), whereas silencing of either WT1 or DNMT1 disrupted this complex with enhanced expression of PAX2. HG-induced DPD dedifferentiation was associated with a higher expression of miR193a, whereas inhibition of miR193a prevented DPD dedifferentiation in HG milieu. HG downregulated DPD expression of APOL1. miR193a-overexpressing DPDs displayed downregulation of APOL1 and enhanced expression of dedifferentiating markers; conversely, silencing of miR193a enhanced the expression of APOL1 and preserved DPD phenotype. Moreover, stably APOL1G0-overexpressing DPDs displayed the enhanced expression of WT1 but attenuated expression of miR193a; nonetheless, silencing of APOL1 reversed these effects. Since silencing of APOL1 enhanced miR193a expression as well as dedifferentiation in DPDs, it appears that downregulation of APOL1 contributed to dedifferentiation of DPDs through enhanced miR193a expression in HG milieu. Vitamin D receptor agonist downregulated miR193a, upregulated APOL1 expression, and prevented dedifferentiation of DPDs in HG milieu. These findings suggest that modulation of the APOL1-miR193a axis carries a potential to preserve DPD molecular phenotype in HG milieu.


Asunto(s)
Apolipoproteína L1/metabolismo , Desdiferenciación Celular/efectos de los fármacos , Glucosa/toxicidad , MicroARNs/metabolismo , Podocitos/efectos de los fármacos , Apolipoproteína L1/genética , Calcitriol/análogos & derivados , Calcitriol/farmacología , Línea Celular Transformada , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo , Fenotipo , Podocitos/metabolismo , Podocitos/patología , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas WT1/genética , Proteínas WT1/metabolismo
6.
Platelets ; 28(5): 521-524, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27885866

RESUMEN

Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder characterized by low platelet counts. First-line treatment includes intravenous immunoglobulin (IVIg), however, its working mechanism remains incompletely understood. We investigated splenic and thymic dendritic cell (DC) subsets upon IVIg treatment in a well-characterized active murine model of ITP. During active disease, there was a significant peripheral deficiency of splenic tolerizing SIRPα+ DCs which could be rescued by IVIg therapy, increasing platelet counts. These splenic tolerizing DC changes were associated with an abrogation of the thymic-retention of tolerizing DCs, suggesting that IVIg may raise platelet counts in ITP by modulating peripheral numbers of tolerizing DCs.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , Púrpura Trombocitopénica Idiopática , Bazo/inmunología , Timo/inmunología , Regulación hacia Arriba/efectos de los fármacos , Animales , Células Dendríticas/patología , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones SCID , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/patología , Bazo/patología , Timo/patología , Regulación hacia Arriba/inmunología
7.
Blood ; 123(3): 422-7, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24258817

RESUMEN

Platelet transfusions are life-saving treatments for many patients with thrombocytopenia; however, their use is generally discouraged in the autoimmune disorder known as immune thrombocytopenia (ITP). We examined whether allogeneic platelet major histocompatibility complex (MHC) class I transfusions affected antiplatelet CD61-induced ITP. BALB/c CD61 knockout mice (CD61(-)/H-2(d)) were immunized against platelets from wild-type syngeneic BALB/c (CD61(+)/H-2(d)), allogeneic C57BL/6 (CD61(+)/H-2(b)), or C57BL/6 CD61 KO (CD61(-)/H-2(b)) mice, and their splenocytes were transferred into severe combined immunodeficient (SCID) mice to induce ITP. When nondepleted splenocytes were transferred to induce antibody-mediated ITP, both CD61(+) platelet immunizations generated immunity that caused thrombocytopenia independently of allogeneic MHC molecules. In contrast, when B-cell-depleted splenocytes were transferred to induce T-cell-mediated ITP, transfer of allogeneic MHC-immunized splenocytes completely prevented CD61-induced ITP development. In addition, allogeneic platelet transfusions into SCID mice with established CD61-induced ITP rescued the thrombocytopenia. Compared with thrombocytopenic mice, bone marrow histology in the rescued mice showed normalized megakaryocyte morphology, and in vitro CD61-specific T-cell cytotoxicity was significantly suppressed. These results indicate that antibody-mediated ITP is resistant to allogeneic platelet transfusions, while the T-cell-mediated form of the disease is susceptible, suggesting that transfusion therapy may be beneficial in antibody-negative ITP.


Asunto(s)
Transfusión de Plaquetas/métodos , Linfocitos T/inmunología , Trombocitopenia/genética , Trombocitopenia/prevención & control , Animales , Células de la Médula Ósea/citología , Modelos Animales de Enfermedad , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina G/inmunología , Integrina beta3/metabolismo , Megacariocitos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Bazo/citología , Trombocitopenia/inmunología , Factores de Tiempo
8.
Blood ; 120(10): 2127-32, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22760780

RESUMEN

Immune thrombocytopenia (ITP) is a bleeding disorder in which antibodies and/or T cells lead to enhanced peripheral platelet destruction and reduced bone marrow platelet production. Several reports have observed that ITP is associated with a peripheral deficiency of tolerance-inducing CD4+CD25+FoxP3+ T regulatory cells (Tregs). Using a murine model of ITP, we analyzed Tregs in the spleen and thymus. CD61 knockout mice were immunized against wild-type (CD61+) platelets, and their splenocytes were transferred into severe combined immunodeficient (SCID) mice. Compared with SCID mice receiving naive splenocytes, within 2 weeks after transfer, the ITP SCID mice became thrombocytopenic (< 200 × 10(9) platelets/L) and had increased serum anti-CD61 antibodies. The quantity of thymic Tregs by 2 weeks after transfer was significantly elevated, whereas Tregs in the spleens were significantly reduced. Treatment of the ITP mice with 2 g/kg intravenous immunoglobulin raised the platelet counts, reduced antibody production, and normalized the thymic and splenic Treg populations. Compared with thymocytes from ITP mice treated with intravenous immunoglobulin, thymocytes from untreated ITP mice delayed the onset of ITP when administered before engraftment with immune splenocytes. These results suggest that ITP in mice is associated with a peripheral Treg deficiency because of thymic retention and therapy normalizes the Tregs.


Asunto(s)
Bazo/inmunología , Linfocitos T Reguladores/inmunología , Trombocitopenia/inmunología , Timo/inmunología , Animales , Anticuerpos/administración & dosificación , Anticuerpos/sangre , Anticuerpos/inmunología , Plaquetas/inmunología , Recuento de Linfocito CD4 , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/inmunología , Enfermedades del Sistema Inmune , Inmunoglobulinas Intravenosas/uso terapéutico , Integrina beta3/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Transfusión de Linfocitos , Ratones , Ratones Noqueados , Ratones SCID , Especificidad de Órganos , Bazo/efectos de los fármacos , Bazo/patología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/patología , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/patología , Timo/efectos de los fármacos , Timo/patología
11.
Diabetes ; 72(12): 1751-1765, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37699387

RESUMEN

Caspases are cysteine-aspartic proteases that were initially discovered to play a role in apoptosis. However, caspase 8, in particular, also has additional nonapoptotic roles, such as in inflammation. Adipocyte cell death and inflammation are hypothesized to be initiating pathogenic factors in type 2 diabetes. Here, we examined the pleiotropic role of caspase 8 in adipocytes and obesity-associated insulin resistance. Caspase 8 expression was increased in adipocytes from mice and humans with obesity and insulin resistance. Treatment of 3T3-L1 adipocytes with caspase 8 inhibitor Z-IETD-FMK decreased both death receptor-mediated signaling and targets of nuclear factor κ-light-chain-enhancer of activated B (NF-κB) signaling. We generated novel adipose tissue and adipocyte-specific caspase 8 knockout mice (aP2Casp8-/- and adipoqCasp8-/-). Both males and females had improved glucose tolerance in the setting of high-fat diet (HFD) feeding. Knockout mice also gained less weight on HFD, with decreased adiposity, adipocyte size, and hepatic steatosis. These mice had decreased adipose tissue inflammation and decreased activation of canonical and noncanonical NF-κB signaling. Furthermore, they demonstrated increased energy expenditure, core body temperature, and UCP1 expression. Adipocyte-specific activation of Ikbkb or housing mice at thermoneutrality attenuated improvements in glucose tolerance. These data demonstrate an important role for caspase 8 in mediating adipocyte cell death and inflammation to regulate glucose and energy homeostasis. ARTICLE HIGHLIGHTS: Caspase 8 is increased in adipocytes from mice and humans with obesity and insulin resistance. Knockdown of caspase 8 in adipocytes protects mice from glucose intolerance and weight gain on a high-fat diet. Knockdown of caspase 8 decreases Fas signaling, as well as canonical and noncanonical nuclear factor κ-light-chain-enhancer of activated B (NF-κB) signaling in adipose tissue. Improved glucose tolerance occurs via reduced activation of NF-κB signaling and via induction of UCP1 in adipocytes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Humanos , Masculino , Femenino , Animales , Ratones , FN-kappa B/metabolismo , Resistencia a la Insulina/genética , Caspasa 8/genética , Caspasa 8/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ratones Noqueados , Adipocitos/metabolismo , Obesidad/genética , Obesidad/metabolismo , Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Glucosa/metabolismo , Apoptosis/genética
12.
Blood ; 116(16): 3073-9, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20616220

RESUMEN

Transfusion-related acute lung injury (TRALI) is a serious complication of transfusion and has been ranked as one of the leading causes of transfusion-related fatalities. Nonetheless, many details of the immunopathogenesis of TRALI, particularly with respect to recipient factors are unknown. We used a murine model of antibody-mediated TRALI in an attempt to understand the role that recipient lymphocytes might play in TRALI reactions. Intravenous injection of an IgG2a antimurine major histocompatibility complex class I antibody (34-1-2s) into BALB/c mice induced moderate hypothermia and pulmonary granulocyte accumulation but no pulmonary edema nor mortality. In contrast, 34-1-2s injections into mice with severe combined immunodeficiency caused severe hypothermia, severe pulmonary edema, and approximately 40% mortality indicating a critical role for T and B lymphocytes in suppressing TRALI reactions. Adoptive transfer of purified CD8(+) T lymphocytes or CD4(+) T cells but not CD19(+) B cells into the severe combined immunodeficiency mice alleviated the antibody-induced hypothermia, lung damage, and mortality, suggesting that T lymphocytes were responsible for the protective effect. Taken together, these results suggest that recipient T lymphocytes play a significant role in suppressing antibody-mediated TRALI reactions. They identify a potentially new recipient mechanism that controls the severity of TRALI reactions.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Anticuerpos/efectos adversos , Linfocitos T/inmunología , Reacción a la Transfusión , Animales , Anticuerpos/inmunología , Quimiocina CXCL2/sangre , Quimiocina CXCL2/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Hipotermia/etiología , Hipotermia/inmunología , Inmunoglobulina G/efectos adversos , Inmunoglobulina G/inmunología , Pulmón/citología , Pulmón/patología , Transfusión de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Neutrófilos/inmunología
13.
Blood ; 115(6): 1247-53, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-20007808

RESUMEN

Immune thrombocytopenia (ITP) is a bleeding disorder characterized by antibody-opsonized platelets being prematurely destroyed in the spleen, although some patients with ITP may have a cell-mediated form of thrombocytopenia. Although several animal models of ITP have been developed, few mimic primary chronic ITP nor have any shown cell-mediated platelet destruction. To create this type of model, splenocytes from CD61 knockout mice immunized against CD61(+) platelets were transferred into severe combined immunodeficient (SCID) (CD61(+)) mouse recipients, and their platelet counts and phenotypes were observed. As few as 5 x 10(4) splenocytes induced a significant thrombocytopenia and bleeding mortality (80%) in recipients within 3 weeks after transfer. Depletion of lymphocyte subsets before transfer showed that the splenocyte's ability to induce thrombocytopenia and bleeding completely depended on CD4(+) T helper cells and that both CD19(+) B cell (antibody)- and CD8(+) T cell (cell)-mediated effector mechanisms were responsible. Treatment of the SCID mouse recipients with intravenous gamma-globulins raised platelet counts and completely prevented bleeding mortality induced by antibody-mediated effector mechanisms but did not affect cell-mediated disease. This novel model not only shows both antibody- and cell-mediated ITP and bleeding but also suggests that these 2 effector mechanisms have a differential response to therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Inmunoglobulinas Intravenosas/administración & dosificación , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/terapia , Animales , Antígenos CD19/inmunología , Plaquetas/inmunología , Femenino , Citometría de Flujo , Integrina beta3/inmunología , Depleción Linfocítica , Megacariocitos/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Recuento de Plaquetas , Púrpura Trombocitopénica Idiopática/patología , Bazo/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología
14.
JCI Insight ; 7(12)2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35511436

RESUMEN

Chronic lymphocytic leukemia (CLL) results from expansion of a CD5+ B cell clone that requires interactions with other cell types, including T cells. Moreover, patients with CLL have elevated levels of circulating IL-17A+ and IL-17F+ CD4+ T (Th17) cells, with higher numbers of IL-17A+ Th17 cells correlating with better outcomes. We report that CLL Th17 cells expressed more miR155, a Th17-differentiation regulator, than control Th17 cells, despite naive CD4+ T (Tn) cell basal miR155 levels being similar in both. We also found that CLL cells directly regulated miR155 levels in Tn cells, thereby affecting Th17 differentiation, by documenting that coculturing Tn cells with resting or activated (Bact) CLL cells altered the magnitude and direction of T cell miR155 levels; CLL Bact cells promoted IL-17A+ and IL-17F+ T cell generation by an miR155-dependent mechanism, confirmed by miR155 inhibition; coculture of Tn cells with CLL Bact cells led to a linear correlation between the degree and direction of T cell miR155 expression changes and production of IL-17F but not IL-17A; and Bact cell-mediated changes in Tn cell miR155 expression correlated with outcome, irrespective of IGHV mutation status, a strong prognostic indicator. These results identify a potentially unrecognized CLL Bact cell-dependent mechanism, upregulation of Tn cell miR155 expression and subsequent enhancement of IL-17F+ Th17 generation, that favors better clinical courses.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , MicroARNs , Células Th17 , Humanos , Interleucina-17/metabolismo , Leucemia Linfocítica Crónica de Células B/genética , MicroARNs/genética , MicroARNs/metabolismo , Células Th17/metabolismo
15.
Mol Metab ; 66: 101594, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36165813

RESUMEN

OBJECTIVE: Adipose tissue is a very dynamic metabolic organ that plays an essential role in regulating whole-body glucose homeostasis. Dysfunctional adipose tissue hypertrophy with obesity is associated with fibrosis and type 2 diabetes. Yes-associated protein 1 (YAP) is a transcription cofactor important in the Hippo signaling pathway. However, the role of YAP in adipose tissue and glucose homeostasis is unknown. METHODS: To study the role of YAP with metabolic stress, we assessed how increased weight and insulin resistance impact YAP in humans and mouse models. To further investigate the in vivo role of YAP specifically in adipose tissue and glucose homeostasis, we developed adipose tissue-specific YAP knockout mice and placed them on either chow or high fat diet (HFD) for 12-14 weeks. To further study the direct role of YAP in adipocytes we used 3T3-L1 cells. RESULTS: We found that YAP protein levels increase in adipose tissue from humans with type 2 diabetes and mouse models of diet-induced obesity and insulin resistance. This suggests that YAP signaling may contribute to adipocyte dysfunction and insulin resistance under metabolic stress conditions. On an HFD, adipose tissue YAP knockout mice had improved glucose tolerance compared to littermate controls. Perigonadal fat pad weight was also decreased in knockout animals, with smaller adipocyte size. Adipose tissue fibrosis and gene expression associated with fibrosis was decreased in vivo and in vitro in 3T3-L1 cells treated with a YAP inhibitor or siRNA. CONCLUSIONS: We show that YAP is increased in adipose tissue with weight gain and insulin resistance. Disruption of YAP in adipocytes prevents glucose intolerance and adipose tissue fibrosis, suggesting that YAP plays an important role in regulating adipose tissue and glucose homeostasis with metabolic stress.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Humanos , Ratones , Animales , Resistencia a la Insulina/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Aumento de Peso , Homeostasis , Fibrosis , Ratones Noqueados , Glucosa/metabolismo
17.
Leukemia ; 35(11): 3163-3175, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33935280

RESUMEN

Cancer pathogenesis involves the interplay of tumor- and microenvironment-derived stimuli. Here we focused on the influence of an immunomodulatory cell type, myeloid-derived suppressor cells (MDSCs), and their lineage-related subtypes on autologous T lymphocytes. Although MDSCs as a group correlated with an immunosuppressive Th repertoire and worse clinical course, MDSC subtypes (polymorphonuclear, PMN-MDSC, and monocytic, M-MDSCs) were often functionally discordant. In vivo, PMN-MDSCs existed in higher numbers, correlated with different Th-subsets, and more strongly associated with poor clinical course than M-MDSCs. In vitro, PMN-MDSCs were more efficient at blocking T-cell growth and promoted Th17 differentiation. Conversely, in vitro M-MDSCs varied in their ability to suppress T-cell proliferation, due to the action of TNFα, and promoted a more immunostimulatory Th compartment. Ibrutinib therapy impacted MDSCs differentially as well, since after initiating therapy, PMN-MDSC numbers progressively declined, whereas M-MDSC numbers were unaffected, leading to a set of less immunosuppressive Th cells. Consistent with this, clinical improvement based on decreasing CLL-cell numbers correlated with the decrease in PMN-MDSCs. Collectively, the data support a balance between PMN-MDSC and M-MDSC numbers and function influencing CLL disease course.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/inmunología , Activación de Linfocitos/inmunología , Células Supresoras de Origen Mieloide/inmunología , Linfocitos T/inmunología , Células TH1/inmunología , Células Th2/inmunología , Microambiente Tumoral , Estudios de Casos y Controles , Diferenciación Celular , Proliferación Celular , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Monocitos/inmunología , Células Supresoras de Origen Mieloide/clasificación , Células Supresoras de Origen Mieloide/patología
18.
Histol Histopathol ; 35(12): 1483-1492, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33124682

RESUMEN

To determine the role of the transplantation of bone marrow-derived mesenchymal stem cells (MSCs) in podocyte renewal, we studied BALB/C mice with or without adriamycin-induced acute kidney injury. MSCs were transplanted ectopically under the capsule of the left kidney or into the peritoneal cavity after the onset of kidney injury to test testing their local or systemic paracrine effects, respectively. Adriamycin produced increases in urine protein: creatinine ratios, blood urea nitrogen, and blood pressure, which improved after both renal subcapsular and intraperitoneal MSCs transplants. The histological changes of adriamycin kidney changes regressed in both kidneys and in only the ipsilateral kidney after intraperitoneal or renal subcapsular transplants indicating that the benefits of transplanted MSCs were related to the extent of paracrine factor distribution. Analysis of kidney tissues for p57-positive parietal epithelial cells (PECs) showed that MSC transplants restored adriamycin-induced decreases in the abundance of these cells to normal levels, although after renal subcapsular transplants these changes did not extend to contralateral kidneys. Moreover, adriamycin caused inflammatory activation of PECs with coexpression of CD44 and phospho-ERK, which was normalized in both or only ipsilateral kidneys depending on whether MSCs were transplanted in the peritoneal cavity or subcapsular space, respectively.


Asunto(s)
Lesión Renal Aguda/cirugía , Proliferación Celular , Trasplante de Células Madre Mesenquimatosas , Podocitos/patología , Regeneración , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Doxorrubicina , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Receptores de Hialuranos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosforilación , Podocitos/metabolismo , Transducción de Señal , Proteínas WT1/metabolismo
19.
Biochimie ; 174: 74-83, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32304771

RESUMEN

EDA2R is a member of the large family of tumor necrosis factor receptor (TNFR). Previous studies suggested that EDA2R expression might be increased in the kidneys of diabetic mice. However, its mRNA and protein expression in kidneys were not analyzed; moreover, its role in the development of diabetic kidney disease was not explored. Here we analyzed the mRNA and protein expressions of EDA2R in diabetic kidneys and examined its role in the podocyte injury in high glucose milieu. By analysis with real-time PCR, Western blotting, we found that both the mRNA and protein levels of EDA2R were increased in the kidneys of diabetic mice. Immunohistochemical studies revealed that EDA2R expression was enhanced in both glomerular and tubular cells of diabetic mice and humans. In vitro studies, high glucose increased EDA2R expression in cultured human podocytes. Overexpression of EDA2R in podocytes promoted podocyte apoptosis and decreased nephrin expression. Moreover, ED2AR increased ROS generation in podocytes, while inhibiting ROS generation attenuates EDA2R-mediated podocyte injury. In addition, EDA2R silencing partially suppressed high glucose-induced ROS generation, apoptosis, and nephrin decrease. Our study demonstrated that high glucose increases EDA2R expression in kidney cells and that EDA2R induces podocyte apoptosis and dedifferentiation in high glucose milieu partially through enhanced ROS generation.


Asunto(s)
Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/metabolismo , Riñón/metabolismo , Podocitos/metabolismo , Receptor Xedar/fisiología , Animales , Apoptosis , Células Cultivadas , Femenino , Riñón/patología , Proteínas de la Membrana/metabolismo , Ratones , Podocitos/patología , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo
20.
Biochimie ; 160: 113-121, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30831151

RESUMEN

BACKGROUND: Increased DAN protein (Grem1, Grem2, Grem3, Cerberus, NBL1, SOST, and USAG1) levels are often associated with severe disease-states in adult kidneys. Grem1, SOST, and USAG1 have been demonstrated to be upregulated and play a critical role in the progression of diabetic nephropathy (DN); however, the expression and the role of other DAN family members in DN have not been reported yet. In this study, we investigated the expression and the role of Grem2 in the development of renal lesions in mice with type 2 DN. METHODS: Fourteen-week-old BTBRob/ob (a mouse model of type 2 diabetes mellitus) and control (BTBR, wild type) mice were evaluated for renal functional and structural biomarkers. Urine was collected for protein content assay, and renal tissues were harvested for molecular analysis with real-time PCR, Western blotting, and immunohistochemistry. In vitro studies, human podocytes were transfected with Grem2 plasmid and were evaluated for apoptosis (morphologic assay and Western blotting). To evaluate the Grem2-mediated downstream signaling, the phosphorylation status of Smad2/3 and Smad1/5/8 was assessed. To establish a causal relationship, the effect of SIS3 (an inhibitor for Samd2/3) and BMP-7 (an agonist for Smad1/5/8) was evaluated on Germ2-induced podocyte apoptosis. RESULTS: BTBRob/ob mice showed elevated urinary protein levels. Renal tissues of BTBRob/ob mice showed an increased expression of Grem2; both glomerular and tubular cells displayed enhanced Grem2 expression. In vitro studies, high glucose increased Grem2 expression in cultured human podocytes, whereas, Grem2 silencing partially protected podocyte from high glucose-induced apoptosis. Overexpression of Grem2 in podocytes not only increased Bax/Bcl2 expression ratio but also promoted podocyte apoptosis; moreover, an overexpression of Grem2 increased the phosphorylation of Smad2/3 and decreased the phosphorylation of Smad1/5/8; furthermore, SIS3 and BMP-7 attenuated Grem2-induced podocyte apoptosis. CONCLUSIONS: High glucose increases Grem2 expression in kidney cells. Grem2 mediates podocyte apoptosis through Smads.


Asunto(s)
Apoptosis , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/patología , Glucosa/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Podocitos/patología , Animales , Citocinas , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Obesos , Fosforilación , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Transducción de Señal , Edulcorantes/farmacología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA