Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 194(7): 1374-1387, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38537932

RESUMEN

Clear-cell renal cell carcinoma (ccRCC), a tubular epithelial malignancy, secretes tumor necrosis factor (TNF), which signals ccRCC cells in an autocrine manner via two cell surface receptors, TNFR1 and TNFR2, to activate shared and distinct signaling pathways. Selective ligation of TNFR2 drives cell cycle entry of malignant cells via a signaling pathway involving epithelial tyrosine kinase, vascular endothelial cell growth factor receptor type 2, phosphatidylinositol-3-kinase, Akt, pSer727-Stat3, and mammalian target of rapamycin. In this study, phosphorylated 4E binding protein-1 (4EBP1) serine 65 (pSer65-4EBP1) was identified as a downstream target of this TNFR2 signaling pathway. pSer65-4EBP1 expression was significantly elevated relative to total 4EBP1 in ccRCC tissue compared with that in normal kidneys, with signal intensity increasing with malignant grade. Selective ligation of TNFR2 with the TNFR2-specific mutein increased pSer65-4EBP1 expression in organ cultures that co-localized with internalized TNFR2 in mitochondria and increased expression of mitochondrially encoded COX (cytochrome c oxidase subunit) Cox1, as well as nuclear-encoded Cox4/5b subunits. Pharmacologic inhibition of mammalian target of rapamycin reduced both TNFR2-specific mutein-mediated phosphorylation of 4EBP1 and cell cycle activation in tumor cells while increasing cell death. These results signify the importance of pSer65-4EBP1 in mediating TNFR2-driven cell-cycle entry in tumor cells in ccRCC and implicate a novel relationship between the TNFR2/pSer65-4EBP1/COX axis and mitochondrial function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Carcinoma de Células Renales , Proteínas de Ciclo Celular , Proliferación Celular , Neoplasias Renales , Mitocondrias , Receptores Tipo II del Factor de Necrosis Tumoral , Transducción de Señal , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Neoplasias Renales/genética , Mitocondrias/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Biosíntesis de Proteínas , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética
2.
Am J Pathol ; 192(4): 722-736, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35063404

RESUMEN

Similar to the behavior of inflamed tubular epithelial cells, clear cell renal cell carcinoma (ccRCC) cells express death receptor 3 (DR3 or TNFSFR25) in situ, and expression increases with tumor grade. Surprisingly, E-selectin, which can be induced in endothelial cells by DR3 signaling, is also expressed by ccRCC cells and increases with tumor grade. In ccRCC organ cultures, addition of tumor necrosis factor-like 1A (TL1A or TNFSF15), the ligand for DR3, activates NF-κB and mitogen-activated protein kinases, induces both DR3 and E-selectin expression in an NF-κB-dependent manner, and promotes cell cycle entry. DR3 immunoprecipitated from ccRCC tissue contains sialyl Lewis X moieties (the ligand recognized by E-selectin), proximity ligation assays reveal DR3, and E-selectin interacts on ccRCC cells. Similar to that with the addition of TL1A, the addition of soluble E-selectin to ccRCC organ cultures activates NF-κB and mitogen-activated protein kinases in ccRCC cells and increases both DR3 and E-selectin expression and cell-cycle entry. In contrast, normal renal tubular epithelium, which poorly expresses DR3, is minimally responsive to either of these ligands. These data suggest a functional role for autocrine/paracrine DR3/E-selectin interactions in ccRCC and its progression, revealing a potential new target for therapeutic intervention.


Asunto(s)
Carcinoma de Células Renales , Selectina E , Neoplasias Renales , Miembro 25 de Receptores de Factores de Necrosis Tumoral , Antígenos CD , Carcinoma de Células Renales/metabolismo , Selectina E/genética , Selectina E/metabolismo , Células Endoteliales/metabolismo , Femenino , Humanos , Neoplasias Renales/metabolismo , Ligandos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
3.
Am J Transplant ; 20(9): 2380-2391, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32167668

RESUMEN

Tumor necrosis factor receptor 2 (TNFR2) is strongly upregulated on renal tubular epithelial cells by acute cell-mediated rejection (ACR. In human kidney organ culture, TNFR2 signaling both upregulates TNFR2 expression and promotes cell cycle entry of tubular epithelial cells. We find significantly more cells express CD133 mRNA and protein, a putative stem cell marker, in allograft biopsy samples with ACR compared to acute tubular injury without rejection or pretransplant "normal kidney" biopsy samples. Of CD133+ cells, ~85% are within injured tubules and ~15% are interstitial. Both populations express stem cell marker TRA-1-60 and TNFR2, but only tubular CD133+ cells express proximal tubular markers megalin and aquaporin-1. TNFR2+ CD133+ cells in tubules express proliferation marker phospho-histone H3S10 (pH3S10 ). Tubular epithelial cells in normal kidney organ cultures respond to TNFR2 signaling by expressing CD133 mRNA and protein, stem cell marker TRA-1-60, and pH3S10 within 3 hours of treatment. This rapid response time suggests that CD133+ cells in regenerating tubules of kidneys undergoing ACR represent proliferating tubular epithelial cells with TNFR2-induced stem cell markers rather than expansion of resident stem cells. Infiltrating host mononuclear cells are a likely source of TNF as these changes are absent in acute tubular injury .


Asunto(s)
Trasplante de Riñón , Neoplasias , Aloinjertos , Células Epiteliales , Rechazo de Injerto/etiología , Humanos , Riñón , Túbulos Renales , Necrosis , Células Madre
4.
J Am Soc Nephrol ; 26(2): 400-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25012180

RESUMEN

Anion exchanger-1 (AE1) mediates chloride-bicarbonate exchange across the plasma membranes of erythrocytes and, via a slightly shorter transcript, kidney epithelial cells. On an omnivorous human diet, kidney AE1 is mainly active basolaterally in α-intercalated cells of the collecting duct, where it is functionally coupled with apical proton pumps to maintain normal acid-base homeostasis. The C-terminal tail of AE1 has an important role in its polarized membrane residency. We have identified the ß1 subunit of Na(+),K(+)-ATPase (sodium pump) as a binding partner for AE1 in the human kidney. Kidney AE1 and ß1 colocalized in renal α-intercalated cells and coimmunoprecipitated (together with the catalytic α1 subunit of the sodium pump) from human kidney membrane fractions. ELISA and fluorescence titration assays confirmed that AE1 and ß1 interact directly, with a Kd value of 0.81 µM. GST-AE1 pull-down assays using human kidney membrane proteins showed that the last 11 residues of AE1 are important for ß1 binding. siRNA-induced knockdown of ß1 in cell culture resulted in a significant reduction in kidney AE1 levels at the cell membrane, whereas overexpression of kidney AE1 increased cell surface sodium pump levels. Notably, membrane staining of ß1 was reduced throughout collecting ducts of AE1-null mouse kidney, where increased fractional excretion of sodium has been reported. These data suggest a requirement of ß1 for proper kidney AE1 membrane residency, and that activities of AE1 and the sodium pump are coregulated in kidney.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/fisiología , Membrana Celular/metabolismo , Riñón/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/deficiencia , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Línea Celular , Membrana Celular/patología , Células Cultivadas , Homeostasis/fisiología , Humanos , Riñón/patología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Unión Proteica , ARN Interferente Pequeño/farmacología , ATPasa Intercambiadora de Sodio-Potasio/efectos de los fármacos
5.
Kidney Int ; 87(2): 281-96, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25140911

RESUMEN

Tumor necrosis factor (TNF), initially reported to induce tumor cell apoptosis and cachexia, is now considered a central mediator of a broad range of biological activities from cell proliferation, cell death and differentiation to induction of inflammation and immune modulation. TNF exerts its biological responses via interaction with two cell surface receptors: TNFR1 and TNFR2. (TNFRs). These receptors trigger shared and distinct signaling pathways upon TNF binding, which in turn result in cellular outputs that may promote tissue injury on one hand but may also induce protective, beneficial responses. Yet the role of TNF and its receptors specifically in renal disease is still not well understood. This review describes the expression of the TNFRs, the signaling pathways induced by them and the biological responses of TNF and its receptors in various animal models of renal diseases, and discusses the current outcomes from use of TNF biologics and TNF biomarkers in renal disorders.


Asunto(s)
Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Animales , Carcinoma de Células Renales/etiología , Carcinoma de Células Renales/metabolismo , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Rechazo de Injerto/etiología , Rechazo de Injerto/metabolismo , Humanos , Enfermedades Renales/terapia , Neoplasias Renales/etiología , Neoplasias Renales/metabolismo , Trasplante de Riñón/efectos adversos , Modelos Biológicos , Nefritis/etiología , Nefritis/metabolismo , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Transducción de Señal , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/metabolismo
6.
Stem Cells ; 31(9): 1881-92, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23712715

RESUMEN

TNF, signaling through TNFR2, has been implicated in tissue repair, a process that in the heart may be mediated by activated resident cardiac stem cells (CSCs). The objective of our study is to determine whether ligation of TNFR2 can induce activation of resident CSCs in the setting of ischemic cardiac injury. We show that in human cardiac tissue affected by ischemia heart disease (IHD), TNFR2 is expressed on intrinsic CSCs, identified as c-kit(+)/CD45(-)/VEGFR2(-) interstitial round cells, which are activated as determined by entry to cell cycle and expression of Lin-28. Wild-type mouse heart organ cultures subjected to hypoxic conditions both increase cardiac TNF expression and show induced TNFR2 and Lin-28 expression in c-kit(+) CSCs that have entered cell cycle. These CSC responses are enhanced by exogenous TNF. TNFR2(-/-) mouse heart organ cultures subjected to hypoxia increase cardiac TNF but fail to induce CSC activation. Similarly, c-kit(+) CSCs isolated from mouse hearts exposed to hypoxia or TNF show induction of Lin-28, TNFR2, cell cycle entry, and cardiogenic marker, α-sarcomeric actin (α-SA), responses more pronounced by hypoxia in combination with TNF. Knockdown of Lin-28 by siRNA results in reduced levels of TNFR2 expression, cell cycle entry, and diminished expression of α-SA. We conclude that hypoxia-induced c-kit(+) CSC activation is mediated by TNF/TNFR2/Lin-28 signaling. These observations suggest that TNFR2 signaling in resident c-kit(+) CSCs induces cardiac repair, findings which provide further understanding of the unanticipated harmful effects of TNF blockade in human IHD.


Asunto(s)
Ciclo Celular , Isquemia Miocárdica/patología , Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Células Madre/citología , Factor de Necrosis Tumoral alfa/metabolismo , Actinas/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Separación Celular , Técnica del Anticuerpo Fluorescente , Humanos , Hibridación in Situ , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Técnicas de Cultivo de Órganos , Proteínas de Unión al ARN/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 33(1): 34-42, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23139294

RESUMEN

OBJECTIVE: Mutations in the bone morphogenetic protein type II receptor (BMPR-II) are responsible for the majority of cases of heritable pulmonary arterial hypertension (PAH), and BMPR-II deficiency contributes to idiopathic and experimental forms of PAH. Sildenafil, a potent type-5 nucleotide-dependent phosphodiesterase inhibitor, is an established treatment for PAH, but whether sildenafil affects bone morphogenetic protein (BMP) signaling in the pulmonary circulation remains unknown. METHODS AND RESULTS: Studies were undertaken in human pulmonary arterial smooth muscle cells (PASMCs) and in vivo in the monocrotaline rat model of PAH. In PASMCs, sildenafil enhanced BMP4-induced phosphorylation of Smad1/5, Smad nuclear localization, and Inhibitor of DNA binding protein 1 gene and protein expression. This effect was mimicked by 8-bromo-cyclic GMP. Pharmacological inhibition or small interfering RNA knockdown of cyclic GMP-dependent protein kinase I inhibited the effect of sildenafil on BMP signaling. In functional studies, we observed that sildenafil potentiated the antiproliferative effects of BMP4 on PASMC proliferation. Furthermore, sildenafil restored the antiproliferative response to BMP4 in PASMCs harboring mutations in BMPR-II. In the monocrotaline rat model of PAH, which is characterized by BMPR-II deficiency, sildenafil prevented the development of pulmonary hypertension and vascular remodeling, and partly restored Smad1/5 phosphorylation and Inhibitor of DNA binding protein 1 gene expression in vivo in monocrotaline exposed rat lungs. CONCLUSIONS: Sildenafil enhances canonical BMP signaling via cyclic GMP and cyclic GMP-dependent protein kinase I in vitro and in vivo, and partly restores deficient BMP signaling in BMPR-II mutant PASMCs. Our findings demonstrate a novel mechanism of action of sildenafil in the treatment of PAH and suggest that targeting BMP signaling may be beneficial in this disease.


Asunto(s)
Antihipertensivos/farmacología , Proteínas Morfogenéticas Óseas/metabolismo , Hipertensión Pulmonar/prevención & control , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Arteria Pulmonar/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sulfonas/farmacología , Vasodilatadores/farmacología , Animales , Sitios de Unión , Proteína Morfogenética Ósea 4/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/efectos de los fármacos , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipertensión Pulmonar Primaria Familiar , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/metabolismo , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Masculino , Monocrotalina , Músculo Liso Vascular/metabolismo , Mutación , Miocitos del Músculo Liso/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Arteria Pulmonar/metabolismo , Purinas/farmacología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Citrato de Sildenafil , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Transfección
8.
BMC Nephrol ; 15: 178, 2014 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-25399326

RESUMEN

BACKGROUND: Death receptors (DRs) play an important role in renal pathology. We have shown that DR3 is inducibly expressed on renal tubular epithelial cells in the setting of inflammatory injuries. In this study we investigate the expression of DR3 in renal endothelial cells and their response to TL1A, the only known ligand of DR3. METHODS: We did RT-PCR, flow cytometry and subcellular immunoblotting to examine the expression and function of DR3 in cells in vitro. We did organ culture of human and mouse tissue to examine expression and signal of DR3 in vivo. RESULTS: DR3 is expressed in some interstitial vascular endothelial cells (EC) in human kidney in situ; these EC also respond to its ligand TL1A by activating NF-κB. Very low levels of DR3 can be detected on the cell surface of cultured human umbilical vein (HUV) EC, which do not respond to TL1A. HUVEC transfected to overexpress DR3 become responsive to TL1A, assessed by IκBα degradation and E-selectin induction, indicating that the signaling components needed for DR3 responsiveness are expressed. TL1A induces NF-κB activation in EC in renal and cardiac tissue from wild type but not DR3 knock-out mice. CONCLUSION: TL1A and DR3 activate NF-κB in vascular endothelial cells, and can be an important regulator of renal interstitial vascular injury.


Asunto(s)
Células Endoteliales/metabolismo , Riñón/citología , FN-kappa B/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas I-kappa B/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/química , Inhibidor NF-kappaB alfa , Técnicas de Cultivo de Órganos , Miembro 25 de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 25 de Receptores de Factores de Necrosis Tumoral/deficiencia , Proteínas Recombinantes/farmacología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología
9.
Am J Physiol Lung Cell Mol Physiol ; 305(4): L312-21, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23771884

RESUMEN

Bone morphogenetic protein type II receptor (BMPR-II) mutations are responsible for over 70% of cases of heritable pulmonary arterial hypertension (PAH). Loss of BMP signaling promotes pulmonary vascular remodeling via modulation of pulmonary artery smooth muscle cell (PASMC) proliferation. Id proteins (Id1-4) are major downstream transcriptional targets of BMP signaling. However, the impact of BMPR-II mutation on the expression of the range of Id proteins and the contribution of individual Id proteins to abnormal PASMC function remain unclear. Human PASMCs were used to determine the expression of Id proteins (Id1-4) by real-time PCR and immunoblotting. The BMP responses in control cells were compared with PASMCs harboring BMPR-II mutations and cells in which BMPR-II was knocked down by siRNA transfection. Id3 expression in pulmonary vessels was also investigated in BMPR-II mutant mice and in patients with heritable PAH. BMP4 and BMP6, but not BMP9, induced mRNA expression of Id1, Id2, and Id3. The BMP-stimulated induction of Id1 and Id3 was markedly reduced in BMPR-II mutant PASMCs and in control PASMCs following siRNA silencing of BMPR-II. Pulmonary arteries in BMPR-II mutant mice and patients with heritable PAH demonstrated reduced levels of Id3 compared with control subjects. Lentiviral overexpression of Id3 reduced cell cycle progression and inhibited proliferation of PASMCs. Lipopolysaccharide further reduced Id3 expression in mutant PASMCs. In conclusion, Id proteins, and particularly Id1 and Id3, are critical downstream effectors of BMP signaling in PASMCs. Loss of BMPR-II function reduces the induction of Id genes in PASMCs, Id1, and Id3 regulate the proliferation of PASMCs via cell cycle inhibition, an effect that may be exacerbated by inflammatory stimuli.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/patología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/deficiencia , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hipertensión Pulmonar Primaria Familiar , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Proteínas Inhibidoras de la Diferenciación/genética , Lipopolisacáridos/farmacología , Pulmón/patología , Ratones , Ratones Mutantes , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
10.
Am J Pathol ; 180(4): 1454-64, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22330679

RESUMEN

The expression of death receptor 3 (DR3), a member of the tumor necrosis factor (TNF) receptor superfamily, is up-regulated in human tubular epithelial cells (TECs) during renal injury, but its function in this setting remains unknown. We used cisplatin to induce renal injury in wild-type (DR3(+/+)) or congenitally deficient DR3(-/-) mice to examine the in vivo role of DR3. Cisplatin induced the expression of DR3, its ligand, TNF-like ligand 1A (TL1A), and TNF in TECs, as observed in human renal injury. Cisplatin increased apoptotic death of DR3(-/-) TECs by twofold compared with DR3(+/+) TECs, whereas it reduced the number of tubules expressing phospho-NF-κBp65(Ser276) by 50% at 72 hours. Similar degrees of induction of DR3, TL1A, and TNF, and changes in apoptosis and phospho-NF-κBp65(Ser276), were obtained in mouse kidney organ cultures treated with cisplatin for 3 hours, suggesting a direct effect on TECs. TNF was implicated in mediating cisplatin-induced tubular damage given that the in vivo co-administration of GM6001, an inhibitor of TNF maturation and release, significantly reduced TNF production and tubular damage. Moreover, TNF exacerbated, whereas TL1A reduced, cisplatin-induced apoptosis in the DR3(+/+) mouse proximal tubule cell line, TKPTS. Our data demonstrate that cisplatin-induced nephrotoxicity is mitigated by DR3 signaling, suggesting that this occurs by antagonizing pro-apoptotic signals induced by TNF. Therefore, activating DR3 may be beneficial in reducing acute kidney injury.


Asunto(s)
Lesión Renal Aguda/patología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Animales , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Dipéptidos/farmacología , Interacciones Farmacológicas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Ligandos , Ratones , Ratones Mutantes , FN-kappa B/metabolismo , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 25 de Receptores de Factores de Necrosis Tumoral/deficiencia , Transducción de Señal/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba/efectos de los fármacos
11.
Arterioscler Thromb Vasc Biol ; 32(2): 353-60, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22053072

RESUMEN

OBJECTIVE: Perioperative nonimmune injuries to an allograft can decrease graft survival. We have developed a model for studying this process using human materials. METHODS AND RESULTS: Human artery segments were transplanted as infrarenal aortic interposition grafts into an immunodeficient mouse host, allowed to "heal in" for 30 days, and then retransplanted into a second mouse host. To induce a reperfusion injury, the healed-in artery segments were incubated for 3 hours under hypoxic conditions ex vivo before retransplantation. To induce immunologic rejection, the animals receiving the retransplanted artery segment were adoptively transferred with human peripheral blood mononuclear cells or purified T cells from a donor allogeneic to the artery 1 week before surgery. To compare rejection of injured versus healthy tissues, these manipulations were combined. Results were analyzed ex vivo by histology, morphometry, immunohistochemistry, and mRNA quantitation or in vivo by ultrasound. Our results showed that reperfusion injury, which otherwise heals with minimal sequelae, intensifies the degree of allogeneic T cell-mediated injury to human artery segments. CONCLUSIONS: We developed a new human-mouse chimeric model demonstrating interactions of reperfusion injury and alloimmunity using human cells and tissues that may be adapted to study other forms of nonimmune injury and other types of adaptive immune responses.


Asunto(s)
Inmunidad Adaptativa/fisiología , Arterias/inmunología , Arterias/trasplante , Quimera/inmunología , Daño por Reperfusión/fisiopatología , Linfocitos T/inmunología , Adulto , Animales , Arterias/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/fisiopatología , Supervivencia de Injerto/inmunología , Supervivencia de Injerto/fisiología , Humanos , Ratones , Ratones SCID , Modelos Animales , Linfocitos T/patología , Trasplante Homólogo
12.
Circ Res ; 107(2): 252-62, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20522807

RESUMEN

RATIONALE: Mutations in the bone morphogenetic protein type II receptor (BMPR-II) are responsible for the majority of cases of heritable pulmonary arterial hypertension (PAH). Mutations lead to reduced Smad1/5-driven expression of inhibitor of DNA binding protein 1 (Id1) and loss of the growth suppressive effects of BMPs. The impact of existing PAH therapies on BMP signaling is lacking. OBJECTIVE: Because prostacyclin analogues are effective treatments for clinical PAH, we hypothesized that these agents enhance Smad1/Id1 signaling. METHODS AND RESULTS: Iloprost alone induced Id1 expression in human pulmonary artery smooth muscle cells (PASMCs), an effect that was independent of Smad1/5 activation but dependent on a cAMP-responsive element in the Id1 promoter. In addition, iloprost and treprostinil enhanced BMP-induced phosphorylation of Smad1/5 and Id1 expression in a cAMP-dependent manner. The mechanism involved suppression of inhibitory Smad, Smad6. Furthermore, iloprost rescued the deficit in Smad1/5 phosphorylation and Id gene expression in PASMCs harboring mutations in BMPR-II and restored growth suppression to BMP4 in mutant PASMCs. We confirmed a critical role for Id1 in PASMC proliferation. Reduced expression of Id1 was observed in concentric intimal lesions of heritable PAH cases. In the monocrotaline rat model of PAH, associated with reduced BMPR-II expression, we confirmed that treprostinil inhibited smooth muscle cell proliferation and prevented progression of PAH while enhancing Smad1/5 phosphorylation and Id1 gene expression. CONCLUSIONS: Prostacyclin analogues enhance Id1 expression in vitro and in vivo and restore deficient BMP signaling in BMPR-II mutant PASMCs.


Asunto(s)
Antihipertensivos/farmacología , Proliferación Celular/efectos de los fármacos , Epoprostenol/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Smad/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Epoprostenol/análogos & derivados , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Iloprost/farmacología , Proteína 1 Inhibidora de la Diferenciación/genética , Masculino , Monocrotalina , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Mutación , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosforilación , Regiones Promotoras Genéticas , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Proteínas Smad/genética , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína smad6/metabolismo , Transfección , Regulación hacia Arriba
13.
Cell Death Dis ; 13(6): 549, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35701409

RESUMEN

Inhibition of DNA binding proteins 1 and 3 (ID1 and ID3) are important downstream targets of BMP signalling that are necessary for embryonic development. However, their specific roles in regulating the pluripotency of human embryonic stem cells (hESCs) remain unclear. Here, we examined the roles of ID1 and ID3 in primed and naive-like hESCs and showed that ID1 and ID3 knockout lines (IDs KO) exhibited decreased survival in both primed and naive-like state. IDs KO lines in the primed state also tended to undergo pluripotent dissolution and ectodermal differentiation. IDs KO impeded the primed-to-naive transition (PNT) of hESCs, and overexpression of ID1 in primed hESCs promoted PNT. Furthermore, single-cell RNA sequencing demonstrated that ID1 and ID3 regulated the survival and pluripotency of hESCs through the AKT signalling pathway. Finally, we showed that TCF3 mediated transcriptional inhibition of MCL1 promotes AKT phosphorylation, which was confirmed by TCF3 knockdown in KO lines. Our study suggests that IDs/TCF3 acts through AKT signalling to promote survival and maintain pluripotency of both primed and naive-like hESCs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células Madre Embrionarias Humanas , Proteína 1 Inhibidora de la Diferenciación , Proteína 2 Inhibidora de la Diferenciación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
14.
iScience ; 25(3): 103971, 2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35224470

RESUMEN

Clotting Factor V (FV) is primarily synthesized in the liver and when cleaved by thrombin forms pro-coagulant Factor Va (FVa). Using whole blood RNAseq and scRNAseq of peripheral blood mononuclear cells, we find that FV mRNA is expressed in leukocytes, and identify neutrophils, monocytes, and T regulatory cells as sources of increased FV in hospitalized patients with COVID-19. Proteomic analysis confirms increased FV in circulating neutrophils in severe COVID-19, and immunofluorescence microscopy identifies FV in lung-infiltrating leukocytes in COVID-19 lung disease. Increased leukocyte FV expression in severe disease correlates with T-cell lymphopenia. Both plasma-derived and a cleavage resistant recombinant FV, but not thrombin cleaved FVa, suppress T-cell proliferation in vitro. Anticoagulants that reduce FV conversion to FVa, including heparin, may have the unintended consequence of suppressing the adaptive immune system.

15.
Am J Pathol ; 177(2): 943-54, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20566746

RESUMEN

Clear cell renal cell carcinoma (ccRCC), a tubular epithelial cell (TEC) malignancy, frequently secretes tumor necrosis factor (TNF). TNF signals via two distinct receptors (TNFRs). TNFR1, expressed in normal kidney primarily on endothelial cells, activates apoptotic signaling kinase 1 and nuclear factor-kappaB (NF-kappaB) and induces cell death, whereas TNFR2, inducibly expressed on endothelial cells and on TECs by injury, activates endothelial/epithelial tyrosine kinase (Etk), which trans-activates vascular endothelial growth factor receptor 2 (VEGFR2) to promote cell proliferation. We investigated TNFR expression in clinical samples and function in short-term organ cultures of ccRCC tissue treated with wild-type TNF or specific muteins selective for TNFR1 (R1-TNF) or TNFR2 (R2-TNF). There is a significant increase in TNFR2 but not TNFR1 expression on malignant TECs that correlates with increasing malignant grade. In ccRCC organ cultures, R1-TNF increases TNFR1, activates apoptotic signaling kinase and NF-kappaB, and promotes apoptosis in malignant TECs. R2-TNF increases TNFR2, activates NF-kappaB, Etk, and VEGFR2 and increases entry into the cell cycle. Wild-type TNF induces both sets of responses. R2-TNF actions are blocked by pretreatment with a VEGFR2 kinase inhibitor. We conclude that TNF, acting through TNFR2, is an autocrine growth factor for ccRCC acting via Etk-VEGFR2 cross-talk, insights that may provide a more effective therapeutic approach to this disease.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Anciano , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Ciclo Celular/fisiología , Activación Enzimática , Femenino , Humanos , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
FASEB Bioadv ; 2(2): 126-144, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32123862

RESUMEN

Clear cell renal cell carcinoma (ccRCC) contains cancer stem-like cells (CSCs) that express CD133 (ccRCC-CD133+). CSCs are rarely in cell cycle and, as nonproliferating cells, resist most chemotherapeutic agents. Previously, we reported that tumor necrosis factor receptor-2 (TNFR2) signaling promotes the cell cycle entry of ccRCC-CD133+CSCs, rendering them susceptible to cell-cycle-dependent chemotherapeutics. Here, we describe a TNFR2-activated signaling pathway in ccRCC-CD133+CSCs that is required for cell survival. Wild-type (wt)TNF or R2TNF but not R1TNF (TNF muteins that selectively bind to TNFR2 and TNFR1) induces phosphorylation of signal transducer and activator of transcription 3 (STAT3) on serine727 but not tyrosine705, resulting in pSTAT3Ser727 translocation to and colocalization with TNFR2 in mitochondria. R2TNF signaling activates a kinase cascade involving the phosphorylation of VEGFR2, PI-3K, Akt, and mTORC. Inhibition of any of the kinases or siRNA knockdown of TNFR2 or STAT3 promotes cell death associated with mitochondrial morphological changes, cytochrome c release, generation of reactive oxygen species, and TUNEL+cells expressing phosphorylated mixed lineage kinase-like (MLKL). Pretreatment with necrostatin-1 is more protective than z-VAD.fmk, suggesting that most death is necroptotic and TNFR2 signaling promotes cell survival by preventing mitochondrial-mediated necroptosis. These data suggest that a TNFR2 selective agonist may offer a potential therapeutic strategy for ccRCC.

17.
Biology (Basel) ; 9(4)2020 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-32272660

RESUMEN

Anti-angiogenic agents, such as the multi-tyrosine kinase inhibitor sunitinib, are key first line therapies for metastatic clear cell renal cell carcinoma (ccRCC), but their mechanism of action is not fully understood. Here, we take steps towards validating a computational prediction based on differential transcriptome network analysis that phosphorylated adapter RNA export protein (PHAX) is associated with sunitinib drug treatment. The regulatory impact factor differential network algorithm run on patient tissue samples suggests PHAX is likely an important regulator through changes in genome-wide network connectivity. Immunofluorescence staining of patient tumours showed strong localisation of PHAX to the microvasculature consistent with the anti-angiogenic effect of sunitinib. In normal kidney tissue, PHAX protein abundance was low but increased with tumour grade (G1 vs. G3/4; p < 0.01), consistent with a possible role in cancer progression. In organ culture, ccRCC cells had higher levels of PHAX protein expression than normal kidney cells, and sunitinib increased PHAX protein expression in a dose dependent manner (untreated vs. 100 µM; p < 0.05). PHAX knockdown in a ccRCC organ culture model impacted the ability of sunitinib to cause cancer cell death (p < 0.0001 untreated vs. treated), suggesting a role for PHAX in mediating the efficacy of sunitinib.

18.
J Am Soc Nephrol ; 19(5): 953-60, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18287561

RESUMEN

Death receptor 3 (DR3), a member of the TNF receptor (TNFR) superfamily, is induced in human renal tubular epithelial cells (TEC) in response to injury. This study examined the expression and actions of TL1A, the principal ligand for DR3. In histologically normal tissue from biopsy or nephrectomy specimens of renal allografts, TL1A mRNA and protein were expressed in vascular endothelial cells but not in TEC. In specimens of acute or antibody-mediated allograft rejection, vascular endothelial cells and infiltrating leukocytes expressed increased TL1A mRNA and protein, but TEC expressed TL1A protein without mRNA, consistent with uptake of exogenous ligand. Addition of TL1A to organ cultures of human or mouse kidney caused activation of NF-kappaB, expression of TNFR2, activation of caspase-3, and apoptosis in TEC. Inhibition of NF-kappaB activation increased TL1A-mediated caspase-3 activation and apoptosis of TEC, but it did not reduce the induction of TNFR2. In organ culture of DR3-deficient mouse kidneys, addition of TL1A induced TNFR2 but did not activate NF-kappaB and did not increase apoptosis of TEC. These data suggest that TL1A may contribute to renal inflammation and injury through DR3-mediated activation of NF-kappaB and caspase-3, respectively, but that an unidentified receptor may mediate the NF-kappaB-independent induction of TNFR2 in TEC.


Asunto(s)
Rechazo de Injerto/fisiopatología , Trasplante de Riñón , Nefritis/fisiopatología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis/fisiología , Biopsia , Caspasa 3/metabolismo , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , FN-kappa B/metabolismo , Nefritis/inmunología , Nefritis/patología , Técnicas de Cultivo de Órganos , ARN Mensajero/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
19.
Sci Rep ; 8(1): 12079, 2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30104686

RESUMEN

Human T regulatory cells (T regs) express high levels of TNF receptor 2 (TNFR2). Ligation of TNFR2 with TNF, which can recognise both TNFR1 and TNFR2, or with a TNFR2-selective binding molecule, DARPin 18 (D18) activates canonical NF-κB signalling, assessed by IκBα degradation, and the magnitude of the response correlates with the level of TNFR2 expression. RNA-seq analysis of TNF- or D18-treated human T regs revealed that TNFR2 ligation induces transcription of NFKB2 and RELB, encoding proteins that form the non-canonical NF-κB transcription factor. In combination with IL2, D18 treatment is specific for T regs in (1) stabilising NF-κB-inducing kinase protein, the activator of non-canonical NF-κB signalling, (2) inducing translocation of RelB from cytosol to nucleus, (3) increasing cell cycle entry, and (4) increasing cell numbers. However, the regulatory function of the expanded T regs is unaltered. Inhibition of RelB nuclear translocation blocks the proliferative response. We conclude that ligation of TNFR2 by D18 enhances IL2-induced T regs proliferation and expansion in cell number through the non-canonical NF-κB pathway.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interleucina-2/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/genética , Linfocitos T Reguladores/inmunología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/genética , Células Cultivadas , Voluntarios Sanos , Humanos , Interleucina-2/inmunología , Inhibidor NF-kappaB alfa/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Cultivo Primario de Células , Proteolisis/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Transcripción ReIB/metabolismo
20.
Front Med (Lausanne) ; 4: 148, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28955710

RESUMEN

Human studies, critical for developing new diagnostics and therapeutics, are limited by ethical and logistical issues, and preclinical animal studies are often poor predictors of human responses. Standard human cell cultures can address some of these concerns but the absence of the normal tissue microenvironment can alter cellular responses. Three-dimensional cultures that position cells on synthetic matrices, or organoid or organ-on-a-chip cultures, in which different cell spontaneously organize contacts with other cells and natural matrix only partly overcome this limitation. Here, we review how human organ cultures (HOCs) can more faithfully preserve in vivo tissue architecture and can better represent disease-associated changes. We will specifically describe how HOCs can be combined with both traditional and more modern morphological techniques to reveal how anatomic location can alter cellular responses at a molecular level and permit comparisons among different cells and different cell types within the same tissue. Examples are provided involving use of HOCs to study inflammation, cancer, and stem cell biology.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA