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











Base de datos
Intervalo de año de publicación
1.
Front Transplant ; 2: 1184620, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38993873

RESUMEN

Background: Donation after circulatory death (DCD) liver allografts are associated with higher rates of primary non-function (PNF) and ischemic cholangiopathy (IC). Advanced recovery techniques, including thoracoabdominal normothermic regional perfusion (TA-NRP), may improve organ utilization and patient and allograft outcomes. Given the increasing US experience with TA-NRP DCD recovery, we evaluated outcomes of DCD liver allografts transplanted after TA-NRP. Methods: Liver allografts transplanted from DCD donors after TA-NRP were identified from 5/1/2021 to 1/31/2022 across 8 centers. Donor data included demographics, functional warm ischemic time (fWIT), total warm ischemia time (tWIT) and total time on TA-NRP. Recipient data included demographics, model of end stage liver disease (MELD) score, etiology of liver disease, PNF, cold ischemic time (CIT), liver function tests, intensive care unit (ICU) and hospital length of stay (LOS), post-operative transplant related complications. Results: The donors' median age was 32 years old and median BMI was 27.4. Median fWIT was 20.5 min; fWIT exceeded 30 min in two donors. Median time to initiation of TA-NRP was 4 min and median time on bypass was 66 min. The median recipient listed MELD and MELD at transplant were 22 and 21, respectively. Median allograft CIT was 292 min. The median length of follow up was 257 days. Median ICU and hospital LOS were 2 and 7 days, respectively. Three recipients required management of anastomotic biliary strictures. No patients demonstrated IC, PNF or required re-transplantation. Conclusion: Liver allografts from TA-NRP DCD donors demonstrated good early allograft and recipient outcomes.

2.
Clin J Am Soc Nephrol ; 17(9): 1293-1304, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35944911

RESUMEN

BACKGROUND AND OBJECTIVES: The etiology of chronic kidney disease of unclear etiology, also known as Mesoamerican nephropathy, remains unclear. We investigated potential etiologies for Mesoamerican nephropathy in an immigrant dialysis population. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Migrants with Mesoamerican nephropathy kidney failure (n=52) were identified by exclusion of known causes of kidney disease and compared using a cross-sectional survey with demographically similar patients with kidney failure from other causes (n=63) and age/sex/place of origin-matched healthy participants (n=16). Survey results were extended to the bench; C57BL/6 mice (n=73) received 10-15 weekly intraperitoneal injections of paraquat (a reactive oxygen species-generating herbicide) or vehicle. Kidney function, histology, and expression of organic cation transporter-2 (proximal tubule entry for paraquat) and multidrug and toxin extrusion 1 (extrusion pathway) were examined. Kidney biopsies from Nicaraguan patients with acute Mesoamerican nephropathy were stained for the above transporters and compared with patients with tubulointerstitial nephritis and without Mesoamerican nephropathy. RESULTS: Patients with Mesoamerican nephropathy and kidney failure were young agricultural workers, almost exclusively men; the majority were from Mexico and El Salvador; and they had prior exposures to agrochemicals, including paraquat (27%). After adjustment for age/sex, exposure to any agrochemical or paraquat was associated with Mesoamerican nephropathy kidney failure (odds ratio, 4.86; 95% confidence interval, 1.82 to 12.96; P=0.002 and odds ratio, 12.25; 95% confidence interval, 1.51 to 99.36; P=0.02, respectively). Adjusted for age/sex and other covariates, 1 year of agrochemical exposure was associated with Mesoamerican nephropathy kidney failure (odds ratio, 1.23; 95% confidence interval, 1.04 to 1.44; P=0.02). Compared with 16 matched healthy controls, Mesoamerican nephropathy kidney failure was significantly associated with exposure to paraquat and agrochemicals. Paraquat-treated male mice developed kidney failure and tubulointerstitial nephritis consistent with Mesoamerican nephropathy. Organic cation transporter-2 expression was higher in male kidneys versus female kidneys. Paraquat treatment increased organic cation transporter-2 expression and decreased multidrug and toxin extrusion 1 expression in male kidneys; similar results were observed in the kidneys of Nicaraguan patients with Mesoamerican nephropathy. CONCLUSIONS: Exposure to agrochemicals is associated with Mesoamerican nephropathy, and chronic exposure of mice to paraquat, a prototypical oxidant, induced kidney failure similar to Mesoamerican nephropathy.


Asunto(s)
Nefritis Intersticial , Insuficiencia Renal Crónica , Insuficiencia Renal , Masculino , Femenino , Animales , Ratones , Paraquat/toxicidad , Estudios Transversales , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/epidemiología , Nefritis Intersticial/patología , Enfermedades Renales Crónicas de Etiología Incierta , Agroquímicos , Cationes
3.
Lab Invest ; 102(5): 534-544, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35046485

RESUMEN

The mitochondrial intracrine Stanniocalcin 1 (STC1) activates mitochondrial anti-oxidant defenses. LRP2 (megalin) shuttles STC1 to the mitochondria through retrograde early endosome-to-Golgi- and Rab32-mediated pathway, and LRP2 KO impairs mitochondrial respiration and glycolysis. We determined STC1-LRP2 interaction domains using HA- and FLAG-tagged fragments of STC1 and LRP2, respectively, co-expressed in HEK293T cells. The trans-membrane domain of LRP2 is required for trafficking to the mitochondria. STC1-FLAG expressed in LRP2 KO cells fails to reach the mitochondria; thus, mitochondrial STC1 is extracellularly-derived via LRP2-mediated trafficking. Tri-leucines L12-14 in LRP2's signal peptide interact with STC1's signal peptide. Mutant LRP2 (L(12-14)A) does not bind STC1, while hSTC1 lacking signal peptide or Leucines L8/9/11 does not bind LRP2. STC1 fails to induce respiration or glycolysis in megalin KO mouse embryonal fibroblasts (MEF) expressing mutant LRP2, while mutant hSTC1 (L8/L9/L11 - > A8/A9/A11) fails to reach the mitochondria or induce respiration and glycolysis in WT MEF. Our data suggest direct regulation of mitochondrial metabolism by extracellular cues and reveal an important role for signal peptides' leucines in protein-protein interactions and mitochondrial biology.


Asunto(s)
Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad , Señales de Clasificación de Proteína , Animales , Glicoproteínas , Células HEK293 , Humanos , Leucina/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Mitocondrias/metabolismo
4.
Transl Res ; 235: 15-31, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33789208

RESUMEN

Studies suggest that biological sex influences susceptibility to kidney diseases with males demonstrating greater risk for developing ischemic acute kidney injury (AKI). Sex-related differences in mitochondrial function and homeostasis exist, likely contributing to sexual dimorphism in kidney injury, but the mechanisms are not well characterized. Our observations reveal lower baseline expression of Sirtuin-3 (Sirt3, a major mitochondrial acetyltransferase) in the kidneys of male mice versus females. We tested the hypothesis that differential expression of kidney Sirt3 may mediate sexual dimorphism in AKI using a bilateral kidney ischemia-reperfusion injury (IRI) model and three transgenic mouse models: (1) mice with global transgenic overexpression of Sirt3; (2) mice with inducible, kidney tubule-specific Sirt3 knockdown (iKD); and (3) mice with global Sirt3 knockout. Low mitochondrial Sirt3 (mtSirt3) in males versus females is associated with development of kidney tubular epithelium vacuoles, increased mitochondrial ROS and susceptibility to IRI. Transgenic overexpression of Sirt3 in males protects against kidney IRI and development of tubular epithelium vacuoles. In both sexes, mice with partial kidney tubular epithelium-specific Sirt3 knockdown display intermediate - while global Sirt3 knockout mice display the highest susceptibility to IRI. Female Sirt3 iKD mice demonstrate decreased survival and kidney function after IRI indistinguishable from control males, abolishing the protective effects observed in females. Mechanistically, observed differences in kidney mtSirt3 are sex hormone-dependent; estradiol increases - while testosterone decreases mtSirt3 protein. Our results demonstrate that Sirt3 is an important contributor to the observed sex-related differences in IRI susceptibility, and a potential therapeutic target in the clinical management of AKI.


Asunto(s)
Riñón/irrigación sanguínea , Daño por Reperfusión/etiología , Sirtuina 3/fisiología , Animales , Estradiol/farmacología , Femenino , Riñón/metabolismo , Masculino , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Caracteres Sexuales , Sirtuina 3/genética , Superóxidos/metabolismo , Testosterona/farmacología
5.
Transpl Int ; 34(4): 612-621, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33545741

RESUMEN

The COVID-19 pandemic has significantly changed the landscape of kidney transplantation in the United States and worldwide. In addition to adversely impacting allograft and patient survival in postkidney transplant recipients, the current pandemic has affected all aspects of transplant care, including transplant referrals and listing, organ donation rates, organ procurement and shipping, and waitlist mortality. Critical decisions were made during this period by transplant centers and individual transplant physicians taking into consideration patient safety and resource utilization. As countries have begun administering the COVID vaccines, new and important considerations pertinent to our transplant population have arisen. This comprehensive review focuses on the impact of COVID-19 on kidney transplantation rates, mortality, policy decisions, and the clinical management of transplanted patients infected with COVID-19.


Asunto(s)
COVID-19 , Política de Salud , Fallo Renal Crónico/cirugía , Trasplante de Riñón/tendencias , Atención Perioperativa/tendencias , Obtención de Tejidos y Órganos/tendencias , Listas de Espera/mortalidad , COVID-19/complicaciones , COVID-19/diagnóstico , COVID-19/epidemiología , COVID-19/prevención & control , Vacunas contra la COVID-19 , Europa (Continente)/epidemiología , Asignación de Recursos para la Atención de Salud , Accesibilidad a los Servicios de Salud/tendencias , Humanos , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/mortalidad , Trasplante de Riñón/métodos , Trasplante de Riñón/mortalidad , Pandemias , Atención Perioperativa/métodos , Obtención de Tejidos y Órganos/métodos , Obtención de Tejidos y Órganos/organización & administración , Estados Unidos/epidemiología
6.
Cancer Cell ; 37(6): 818-833.e9, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32516591

RESUMEN

T cells are central to all currently effective cancer immunotherapies, but the characteristics defining therapeutically effective anti-tumor T cells have not been comprehensively elucidated. Here, we delineate four phenotypic qualities of effective anti-tumor T cells: cell expansion, differentiation, oxidative stress, and genomic stress. Using a CRISPR-Cas9-based genetic screen of primary T cells we measured the multi-phenotypic impact of disrupting 25 T cell receptor-driven kinases. We identified p38 kinase as a central regulator of all four phenotypes and uncovered transcriptional and antioxidant pathways regulated by p38 in T cells. Pharmacological inhibition of p38 improved the efficacy of mouse anti-tumor T cells and enhanced the functionalities of human tumor-reactive and gene-engineered T cells, paving the way for clinically relevant interventions.


Asunto(s)
Neoplasias de la Mama/terapia , Sistemas CRISPR-Cas , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Fenotipo , Linfocitos T/trasplante , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Diferenciación Celular , Femenino , Ingeniería Genética , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
7.
N Engl J Med ; 382(5): 416-426, 2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-31995687

RESUMEN

BACKGROUND: Acute kidney injury is common, with a major effect on morbidity and health care utilization. Soluble urokinase plasminogen activator receptor (suPAR) is a signaling glycoprotein thought to be involved in the pathogenesis of kidney disease. We investigated whether a high level of suPAR predisposed patients to acute kidney injury in multiple clinical contexts, and we used experimental models to identify mechanisms by which suPAR acts and to assess it as a therapeutic target. METHODS: We measured plasma levels of suPAR preprocedurally in patients who underwent coronary angiography and patients who underwent cardiac surgery and at the time of admission to the intensive care unit in critically ill patients. We assessed the risk of acute kidney injury at 7 days as the primary outcome and acute kidney injury or death at 90 days as a secondary outcome, according to quartile of suPAR level. In experimental studies, we used a monoclonal antibody to urokinase plasminogen activator receptor (uPAR) as a therapeutic strategy to attenuate acute kidney injury in transgenic mice receiving contrast material. We also assessed cellular bioenergetics and generation of reactive oxygen species in human kidney proximal tubular (HK-2) cells that were exposed to recombinant suPAR. RESULTS: The suPAR level was assessed in 3827 patients who were undergoing coronary angiography, 250 who were undergoing cardiac surgery, and 692 who were critically ill. Acute kidney injury developed in 318 patients (8%) who had undergone coronary angiography. The highest suPAR quartile (vs. the lowest) had an adjusted odds ratio of 2.66 (95% confidence interval [CI], 1.77 to 3.99) for acute kidney injury and 2.29 (95% CI, 1.71 to 3.06) for acute kidney injury or death at 90 days. Findings were similar in the surgical and critically ill cohorts. The suPAR-overexpressing mice that were given contrast material had greater functional and histologic evidence of acute kidney injury than wild-type mice. The suPAR-treated HK-2 cells showed heightened energetic demand and mitochondrial superoxide generation. Pretreatment with a uPAR monoclonal antibody attenuated kidney injury in suPAR-overexpressing mice and normalized bioenergetic changes in HK-2 cells. CONCLUSIONS: High suPAR levels were associated with acute kidney injury in various clinical and experimental contexts. (Funded by the National Institutes of Health and others.).


Asunto(s)
Lesión Renal Aguda/sangre , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Angiografía Coronaria/efectos adversos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Anciano , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Biomarcadores/sangre , Enfermedad Crítica , Modelos Animales de Enfermedad , Femenino , Humanos , Unidades de Cuidados Intensivos , Túbulos Renales/citología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Oportunidad Relativa , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/etiología , Medición de Riesgo/métodos , Activador de Plasminógeno de Tipo Uroquinasa/farmacología
8.
Med Res Arch ; 7(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-31276028

RESUMEN

The kidney is one of the most energy-demanding organs in the human body, and the maintenance of mitochondrial homeostasis is central to kidney function. Recent advances have led to a greater appreciation of how mitochondrial dysfunction contributes to the pathogenesis of AKI, from decreased ATP production, to enhanced mitochondrial oxidative stress, cell necrosis and apoptosis. Accumulating evidence suggests sexual dimorphism in the response to AKI with males demonstrating greater risk for developing ischemia-reperfusion and sepsis-induced kidney injury. In contrast, females may be more susceptible to nephrotoxic-AKI. There are important sex-related differences in mitochondrial respiration, biogenesis and dynamics that likely contribute to the observed sexual dimorphism in AKI. Sex hormones mediate many of these differences with multiple preclinical studies demonstrating the renoprotective actions of estrogen in many rodent models of AKI. Estrogenic control of mitochondrial biogenesis, function and reactive oxygen species (ROS) generation is discussed. Furthermore, the potential role for sex chromosomes in mediating sex differences in AKI is examined. Novel animal models such as the "four core genotypes" (FCG) mouse model provide us with important tools to study sex chromosome effects in kidney health and disease. By understanding the influences of sexual dimorphism or sex hormones on mitochondrial homeostasis and disease manifestations, we may be able to identify novel therapeutic targets and improve existing treatment options for AKI.

9.
Trauma Surg Acute Care Open ; 4(1): e000334, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31321313

RESUMEN

CASE PRESENTATION: A 38-year-old man was brought in by ambulance as a trauma activation after sustaining a self-inflicted stab wound in the left upper quadrant with a kitchen knife. His primary survey was unremarkable and his vital signs were normal. Secondary survey revealed a 2 cm transverse stab wound inferior and medial to the left nipple. Extended focused assessment with sonography for trauma (FAST) did not show intra-abdominal or pericardial fluid and chest X-ray did not show a definite pneumothorax or hemothorax. WHAT WOULD YOU DO?: Wound exploration at bedside.Admit for observation and serial examinations.Exploratory laparotomy and open repair of traumatic diaphragmatic injury (TDI).Thoracotomy and open repair of TDI.Diagnostic laparoscopy and laparoscopic repair of TDI.

10.
Am J Nephrol ; 48(6): 472-481, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30472701

RESUMEN

BACKGROUND: Death with graft function remains an important cause of graft loss among kidney transplant recipients (KTRs). Little is known about the trend of specific causes of death in KTRs in recent years. METHODS: We analyzed United States Renal Data System data (1996-2014) to determine 1- and 10-year all-cause and cause-specific mortality in adult KTRs who died with a functioning allograft. We also studied 1- and 10-year trends in the various causes of mortality. RESULTS: Of 210,327 KTRs who received their first kidney transplant from 1996 to 2014, 3.2% died within 1 year after transplant. Cardiovascular deaths constituted the majority (24.7%), followed by infectious (15.2%) and malignant (2.9%) causes; 40.1% of deaths had no reported cause. Using 1996 as the referent year, all-cause as well as cardiovascular mortality declined, whereas mortality due to malignancy did not. For analyses of 10-year mortality, we studied 94,384 patients who received a first kidney transplant from 1996 to 2005. Of those, 22.1% died over 10 years and the causative patterns of their causes of death were similar to those associated with 1-year mortality. CONCLUSIONS: Despite the downtrend in mortality over the last 2 decades, a significant percentage of KTRs die in 10-years with a functioning graft, and cardiovascular mortality remains the leading cause of death. These data also highlight the need for diligent collection of mortality data in KTRs.


Asunto(s)
Enfermedades Cardiovasculares/mortalidad , Causas de Muerte/tendencias , Supervivencia de Injerto , Fallo Renal Crónico/mortalidad , Trasplante de Riñón , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Enfermedades Cardiovasculares/etiología , Estudios de Cohortes , Femenino , Humanos , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/cirugía , Masculino , Persona de Mediana Edad , Sistema de Registros/estadística & datos numéricos , Factores de Tiempo , Receptores de Trasplantes/estadística & datos numéricos , Estados Unidos/epidemiología , Adulto Joven
11.
Front Med (Lausanne) ; 5: 211, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30109232

RESUMEN

The risk of mineral and bone disorders among patients with chronic kidney disease is substantially elevated, owing largely to alterations in calcium, phosphorus, vitamin D, parathyroid hormone, and fibroblast growth factor 23. The interwoven relationship among these minerals and hormones results in maladaptive responses that are differentially affected by the process of kidney transplantation. Interpretation of conventional labs, imaging, and other fracture risk assessment tools are not standardized in the post-transplant setting. Post-transplant bone disease is not uniformly improved and considerable variation exists in monitoring and treatment practices. A spectrum of abnormalities such as hypophosphatemia, hypercalcemia, hyperparathyroidism, osteomalacia, osteopenia, and osteoporosis are commonly encountered in the post-transplant period. Thus, reducing fracture risk and other bone-related complications requires recognition of these abnormalities along with the risk incurred by concomitant immunosuppression use. As kidney transplant recipients continue to age, the drivers of bone disease vary throughout the post-transplant period among persistent hyperparathyroidism, de novo hyperparathyroidism, and osteoporosis. The use of anti-resorptive therapies require understanding of different options and the clinical scenarios that warrant their use. With limited studies underscoring clinical events such as fractures, expert understanding of MBD physiology, and surrogate marker interpretation is needed to determine ideal and individualized therapy.

12.
Cell Mol Life Sci ; 75(21): 4021-4040, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29916093

RESUMEN

Mitochondrial intracrines are extracellular signaling proteins, targeted to the mitochondria. The pathway for mitochondrial targeting of mitochondrial intracrines and actions in the mitochondria remains unknown. Megalin/LRP2 mediates the uptake of vitamins and proteins, and is critical for clearance of amyloid-ß protein from the brain. Megalin mutations underlie the pathogenesis of Donnai-Barrow and Lowe syndromes, characterized by brain defects and kidney dysfunction; megalin was not previously known to reside in the mitochondria. Here, we show megalin is present in the mitochondria and associates with mitochondrial anti-oxidant proteins SIRT3 and stanniocalcin-1 (STC1). Megalin shuttles extracellularly-applied STC1, angiotensin II and TGF-ß to the mitochondria through the retrograde early endosome-to-Golgi transport pathway and Rab32. Megalin knockout in cultured cells impairs glycolytic and respiratory capacities. Thus, megalin is critical for mitochondrial biology; mitochondrial intracrine signaling is a continuum of the retrograde early endosome-to-Golgi-Rab32 pathway and defects in this pathway may underlie disease processes in many systems.


Asunto(s)
Péptidos beta-Amiloides/genética , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Mitocondrias/genética , Proteínas de Unión al GTP rab/genética , Agenesia del Cuerpo Calloso/genética , Agenesia del Cuerpo Calloso/metabolismo , Agenesia del Cuerpo Calloso/patología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Membrana Celular/genética , Glicoproteínas/genética , Células HEK293 , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/patología , Hernias Diafragmáticas Congénitas/genética , Hernias Diafragmáticas Congénitas/metabolismo , Hernias Diafragmáticas Congénitas/patología , Humanos , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Mitocondrias/metabolismo , Miopía/genética , Miopía/metabolismo , Miopía/patología , Síndrome Oculocerebrorrenal/genética , Síndrome Oculocerebrorrenal/metabolismo , Síndrome Oculocerebrorrenal/patología , Proteinuria/genética , Proteinuria/metabolismo , Proteinuria/patología , Células RAW 264.7 , Defectos Congénitos del Transporte Tubular Renal/genética , Defectos Congénitos del Transporte Tubular Renal/metabolismo , Defectos Congénitos del Transporte Tubular Renal/patología , Transducción de Señal , Sirtuina 3/genética , Factor de Crecimiento Transformador beta/genética , Proteínas de Unión al GTP rab/metabolismo
13.
Nature ; 537(7621): 539-543, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27626381

RESUMEN

Tumours progress despite being infiltrated by tumour-specific effector T cells. Tumours contain areas of cellular necrosis, which are associated with poor survival in a variety of cancers. Here, we show that necrosis releases intracellular potassium ions into the extracellular fluid of mouse and human tumours, causing profound suppression of T cell effector function. Elevation of the extracellular potassium concentration ([K+]e) impairs T cell receptor (TCR)-driven Akt-mTOR phosphorylation and effector programmes. Potassium-mediated suppression of Akt-mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A. Although the suppressive effect mediated by elevated [K+]e is independent of changes in plasma membrane potential (Vm), it requires an increase in intracellular potassium ([K+]i). Accordingly, augmenting potassium efflux in tumour-specific T cells by overexpressing the potassium channel Kv1.3 lowers [K+]i and improves effector functions in vitro and in vivo and enhances tumour clearance and survival in melanoma-bearing mice. These results uncover an ionic checkpoint that blocks T cell function in tumours and identify potential new strategies for cancer immunotherapy.


Asunto(s)
Cationes Monovalentes/metabolismo , Melanoma/inmunología , Potasio/metabolismo , Linfocitos T/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Humanos , Tolerancia Inmunológica/inmunología , Inmunoterapia/métodos , Canal de Potasio Kv1.3/metabolismo , Masculino , Melanoma/metabolismo , Melanoma/patología , Melanoma/terapia , Potenciales de la Membrana , Ratones , Necrosis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Análisis de Supervivencia , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
14.
Cell ; 166(5): 1117-1131.e14, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565342

RESUMEN

Cancer cells must evade immune responses at distant sites to establish metastases. The lung is a frequent site for metastasis. We hypothesized that lung-specific immunoregulatory mechanisms create an immunologically permissive environment for tumor colonization. We found that T-cell-intrinsic expression of the oxygen-sensing prolyl-hydroxylase (PHD) proteins is required to maintain local tolerance against innocuous antigens in the lung but powerfully licenses colonization by circulating tumor cells. PHD proteins limit pulmonary type helper (Th)-1 responses, promote CD4(+)-regulatory T (Treg) cell induction, and restrain CD8(+) T cell effector function. Tumor colonization is accompanied by PHD-protein-dependent induction of pulmonary Treg cells and suppression of IFN-γ-dependent tumor clearance. T-cell-intrinsic deletion or pharmacological inhibition of PHD proteins limits tumor colonization of the lung and improves the efficacy of adoptive cell transfer immunotherapy. Collectively, PHD proteins function in T cells to coordinate distinct immunoregulatory programs within the lung that are permissive to cancer metastasis. PAPERCLIP.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Pulmón/inmunología , Oxígeno/metabolismo , Prolil Hidroxilasas/metabolismo , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/enzimología , Glucólisis/inmunología , Interferón gamma/inmunología , Pulmón/patología , Neoplasias Pulmonares/terapia , Activación de Linfocitos , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neuropilina-1/metabolismo , Prolil Hidroxilasas/genética , Linfocitos T Reguladores/enzimología , Células TH1/enzimología , Células TH1/inmunología
15.
Nat Immunol ; 17(7): 851-860, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27158840

RESUMEN

T cell antigen receptor (TCR) signaling drives distinct responses depending on the differentiation state and context of CD8(+) T cells. We hypothesized that access of signal-dependent transcription factors (TFs) to enhancers is dynamically regulated to shape transcriptional responses to TCR signaling. We found that the TF BACH2 restrains terminal differentiation to enable generation of long-lived memory cells and protective immunity after viral infection. BACH2 was recruited to enhancers, where it limited expression of TCR-driven genes by attenuating the availability of activator protein-1 (AP-1) sites to Jun family signal-dependent TFs. In naive cells, this prevented TCR-driven induction of genes associated with terminal differentiation. Upon effector differentiation, reduced expression of BACH2 and its phosphorylation enabled unrestrained induction of TCR-driven effector programs.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Linfocitos T CD8-positivos/fisiología , Factor de Transcripción AP-1/metabolismo , Virus Vaccinia/inmunología , Vaccinia/inmunología , Inmunidad Adaptativa , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Linfocitos T CD8-positivos/virología , Diferenciación Celular/genética , Células Cultivadas , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Memoria Inmunológica/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Oncogénica p65(gag-jun) , Transducción de Señal/genética , Factor de Transcripción AP-1/genética
16.
J Clin Invest ; 126(2): 599-604, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26731475

RESUMEN

The immune system has a powerful ability to recognize and kill cancer cells, but its function is often suppressed within tumors, preventing clearance of disease. Functionally diverse innate and adaptive cellular lineages either drive or constrain immune reactions within tumors. The transcription factor (TF) BACH2 regulates the differentiation of multiple innate and adaptive cellular lineages, but its role in controlling tumor immunity has not been elucidated. Here, we demonstrate that BACH2 is required to establish immunosuppression within tumors. Tumor growth was markedly impaired in Bach2-deficient mice and coincided with intratumoral activation of both innate and adaptive immunity. However, augmented tumor clearance in the absence of Bach2 was dependent upon the adaptive immune system. Analysis of tumor-infiltrating lymphocytes from Bach2-deficient mice revealed high frequencies of rapidly proliferating effector CD4+ and CD8+ T cells that expressed the inflammatory cytokine IFN-γ. Effector T cell activation coincided with a reduction in the frequency of intratumoral Foxp3+ Tregs. Mechanistically, BACH2 promoted tumor immunosuppression through Treg-mediated inhibition of intratumoral CD8+ T cells and IFN-γ. These findings demonstrate that BACH2 is a key component of the molecular program of tumor immunosuppression and identify therapeutic targets for the reversal of immunosuppression in cancer.


Asunto(s)
Inmunidad Adaptativa , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Innata , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Linfocitos T CD8-positivos/patología , Interferón gamma/genética , Interferón gamma/inmunología , Ratones , Ratones Noqueados , Neoplasias/genética , Neoplasias/patología , Linfocitos T Reguladores/patología
17.
Toxins (Basel) ; 8(1)2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26712788

RESUMEN

In traditional medicine, Carica papaya leaf has been used for a wide range of therapeutic applications including skin diseases and cancer. In this study, we investigated the in vitro cytotoxicity of aqueous and ethanolic extracts of Carica papaya leaves on the human oral squamous cell carcinoma SCC25 cell line in parallel with non-cancerous human keratinocyte HaCaT cells. Two out of four extracts showed a significantly selective effect towards the cancer cells and were found to contain high levels of phenolic and flavonoid compounds. The chromatographic and mass spectrometric profiles of the extracts obtained with Ultra High Performance Liquid Chromatography-Quadrupole Time of Flight-Mass Spectrometry were used to tentatively identify the bioactive compounds using comparative analysis. The principal compounds identified were flavonoids or flavonoid glycosides, particularly compounds from the kaempferol and quercetin families, of which several have previously been reported to possess anticancer activities. These results confirm that papaya leaf is a potential source of anticancer compounds and warrant further scientific investigation to validate the traditional use of papaya leaf to treat cancer.


Asunto(s)
Antineoplásicos/farmacología , Carica , Extractos Vegetales/farmacología , Antineoplásicos/química , Carcinoma de Células Escamosas , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Flavonoides/análisis , Humanos , Espectrometría de Masas , Fenoles/análisis , Extractos Vegetales/química , Hojas de la Planta
18.
Sci Rep ; 5: 18117, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26640170

RESUMEN

Thrombin-induced and proteinase-activated receptor 1 (PAR1)-mediated signaling increases ROS production, activates ERK, and promotes inflammation and fibroblast proliferation in bleomycin-induced lung injury. Stanniocalcin-1 (STC1) activates anti-oxidant pathways, inhibits inflammation and provides cytoprotection; hence, we hypothesized that STC1 will inhibit thrombin/PAR1 signaling and protect from bleomycin-induced pneumonitis. We determined thrombin level and activity, thrombin-induced PAR-1-mediated signaling, superoxide generation and lung pathology after intra-tracheal administration of bleomycin to WT and STC1 Tg mice. Lungs of bleomycin-treated WT mice display: severe pneumonitis; increased generation of superoxide; vascular leak; increased thrombin protein abundance and activity; activation of ERK; greater cytokine/chemokine release and infiltration with T-cells and macrophages. Lungs of STC1 Tg mice displayed none of the above changes. Mechanistic analysis in cultured pulmonary epithelial cells (A549) suggests that STC1 inhibits thrombin-induced and PAR1-mediated ERK activation through suppression of superoxide. In conclusion, STC1 blunts bleomycin-induced rise in thrombin protein and activity, diminishes thrombin-induced signaling through PAR1 to ERK, and inhibits bleomycin-induced pneumonitis. Moreover, our study identifies a new set of cytokines/chemokines, which play a role in the pathogenesis of bleomycin-induced lung injury. These findings broaden the array of potential therapeutic targets for the treatment of lung diseases characterized by thrombin activation, oxidant stress and inflammation.


Asunto(s)
Glicoproteínas/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Transducción de Señal , Trombina/metabolismo , Animales , Bleomicina , Línea Celular , Quimiocina CXCL16 , Quimiocina CXCL6/metabolismo , Quimiocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glicoproteínas/sangre , Humanos , Mediadores de Inflamación/metabolismo , Lesión Pulmonar/sangre , Lesión Pulmonar/complicaciones , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteopontina/metabolismo , Fosforilación/efectos de los fármacos , Neumonía/sangre , Neumonía/inducido químicamente , Neumonía/complicaciones , Neumonía/metabolismo , Neumonía/patología , Sustancias Protectoras , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Superóxidos/metabolismo
20.
PLoS One ; 10(9): e0138440, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26393521

RESUMEN

BACKGROUND: Inflammation is the hallmark of nephrotoxic nephritis. Stanniocalcin-1 (STC1), a pro-survival factor, inhibits macrophages, stabilizes endothelial barrier function, and diminishes trans-endothelial migration of leukocytes; consistently, transgenic (Tg) overexpression of STC1 protects from nephrotoxic nephritis. Herein, we sought to determine the phenotype of nephrotoxic nephritis after conditional and kidney-specific knockdown of STC1. METHODS: We used Tg mice that, express either STC1 shRNA (70% knockdown of STC1 within 4d) or scrambled shRNA (control) upon delivery of Cre-expressing plasmid to the kidney using ultrasound microbubble technique. Sheep anti-mouse GBM antibody was administered 4d after shRNA activation; and mice were euthanized 10 days later for analysis. RESULTS: Serum creatinine, proteinuria, albuminuria and urine output were similar 10 days after anti-GBM delivery in both groups; however, anti-GBM antibody delivery to mice with kidney-specific knockdown of STC1 produced severe nephrotoxic nephritis, characterized by severe tubular necrosis, glomerular hyalinosis/necrosis and massive cast formation, while control mice manifested mild tubular injury and crescentic glomerulonephritis. Surprisingly, the expression of cytokines/chemokines and infiltration with T-cells and macrophages were also diminished in STC1 knockdown kidneys. Staining for sheep anti-mouse GBM antibody, deposition of mouse C3 and IgG in the kidney, and antibody response to sheep IgG were equal. CONCLUSIONS: nephrotoxic nephritis after kidney-specific knockdown of STC1 is characterized by severe tubular and glomerular necrosis, possibly due to loss of STC1-mediated pro-survival factors, and we attribute the paucity of inflammation to diminished release of cytokines/chemokines/growth factors from the necrotic epithelium.


Asunto(s)
Glicoproteínas/metabolismo , Riñón/metabolismo , Nefritis/etiología , Actinas/metabolismo , Albúminas/análisis , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Línea Celular , Creatinina/sangre , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/genética , Peróxido de Hidrógeno/toxicidad , Inmunoglobulina G/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nefritis/metabolismo , Nefritis/patología , Fenotipo , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA