Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Trends Immunol ; 43(9): 757-775, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35965153

RESUMEN

The procurement and management of nutrients and ability to fight infections are fundamental requirements for survival. These defense responses are bioenergetically costly, requiring the immune system to balance protection against pathogens with the need to maintain metabolic homeostasis. NF-κB transcription factors are central regulators of immunity and inflammation. Over the last two decades, these factors have emerged as a pivotal node coordinating the immune and metabolic systems in physiology and the etiopathogenesis of major threats to human health, including cancer, autoimmunity, chronic inflammation, and others. In this review, we discuss recent advances in understanding how NF-κB-dependent metabolic programs control inflammation, metabolism, and immunity and how improved knowledge of them may lead to better diagnostics and therapeutics for widespread human diseases.


Asunto(s)
FN-kappa B , Neoplasias , Autoinmunidad , Homeostasis , Humanos , Inflamación
2.
Mol Ther ; 31(12): 3531-3544, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37805713

RESUMEN

In vivo apoptosis of human mesenchymal stromal cells (MSCs) plays a critical role in delivering immunomodulation. Yet, caspase activity not only mediates the dying process but also death-independent functions that may shape the immunogenicity of apoptotic cells. Therefore, a better characterization of the immunological profile of apoptotic MSCs (ApoMSCs) could shed light on their mechanistic action and therapeutic applications. We analyzed the transcriptomes of MSCs undergoing apoptosis and identified several immunomodulatory factors and chemokines dependent on caspase activation following Fas stimulation. The ApoMSC secretome inhibited human T cell proliferation and activation, and chemoattracted monocytes in vitro. Both immunomodulatory activities were dependent on the cyclooxygenase2 (COX2)/prostaglandin E2 (PGE2) axis. To assess the clinical relevance of ApoMSC signature, we used the peripheral blood mononuclear cells (PBMCs) from a cohort of fistulizing Crohn's disease (CD) patients who had undergone MSC treatment (ADMIRE-CD). Compared with healthy donors, MSCs exposed to patients' PBMCs underwent apoptosis and released PGE2 in a caspase-dependent manner. Both PGE2 and apoptosis were significantly associated with clinical responses to MSCs. Our findings identify a new mechanism whereby caspase activation delivers ApoMSC immunosuppression. Remarkably, such molecular signatures could implicate translational tools for predicting patients' clinical responses to MSC therapy in CD.


Asunto(s)
Enfermedad de Crohn , Células Madre Mesenquimatosas , Humanos , Enfermedad de Crohn/genética , Enfermedad de Crohn/terapia , Dinoprostona/metabolismo , Leucocitos Mononucleares/metabolismo , Secretoma , Células Madre Mesenquimatosas/metabolismo , Inmunomodulación , Apoptosis , Caspasas
3.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33883278

RESUMEN

Cancer cells can survive chemotherapy-induced stress, but how they recover from it is not known. Using a temporal multiomics approach, we delineate the global mechanisms of proteotoxic stress resolution in multiple myeloma cells recovering from proteasome inhibition. Our observations define layered and protracted programs for stress resolution that encompass extensive changes across the transcriptome, proteome, and metabolome. Cellular recovery from proteasome inhibition involved protracted and dynamic changes of glucose and lipid metabolism and suppression of mitochondrial function. We demonstrate that recovering cells are more vulnerable to specific insults than acutely stressed cells and identify the general control nonderepressable 2 (GCN2)-driven cellular response to amino acid scarcity as a key recovery-associated vulnerability. Using a transcriptome analysis pipeline, we further show that GCN2 is also a stress-independent bona fide target in transcriptional signature-defined subsets of solid cancers that share molecular characteristics. Thus, identifying cellular trade-offs tied to the resolution of chemotherapy-induced stress in tumor cells may reveal new therapeutic targets and routes for cancer therapy optimization.


Asunto(s)
Neoplasias/tratamiento farmacológico , Estrés Fisiológico/efectos de los fármacos , Antineoplásicos/farmacología , Autofagia/fisiología , Línea Celular Tumoral , Humanos , Metaboloma/genética , Mitocondrias/metabolismo , Mieloma Múltiple/metabolismo , Neoplasias/metabolismo , Neoplasias/fisiopatología , Inhibidores de Proteasoma/farmacología , Proteolisis , Proteoma/genética , Análisis de Sistemas , Transcriptoma/genética
4.
Semin Cell Dev Biol ; 98: 118-128, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31132468

RESUMEN

NF-κB plays a pivotal role in oncogenesis. This transcription factor is best known for promoting cancer cell survival and tumour-driving inflammation. However, several lines of evidence support a crucial role for NF-κB in governing energy homeostasis and mediating cancer metabolic reprogramming. Mitochondria are central players in many metabolic processes altered in cancer. Beyond their bioenergetic activity, several facets of mitochondria biology, including mitochondrial dynamics and oxidative stress, promote and sustain malignant transformation. Recent reports revealed an intimate connection between NF-κB pathway and the oncogenic mitochondrial functions. NF-κB can impact mitochondrial respiration and mitochondrial dynamics, and, reciprocally, mitochondria can sense stress signals and convert them into cell biological responses leading to NF-κB activation. In this review we discuss their emerging reciprocal regulation and the significance of this interplay for anticancer therapy.


Asunto(s)
Mitocondrias/metabolismo , FN-kappa B/metabolismo , Neoplasias/metabolismo , Animales , Antineoplásicos/farmacología , Reprogramación Celular/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología
6.
Genes (Basel) ; 15(2)2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38397187

RESUMEN

Tumor-associated macrophages (TAMs) are the major component of the tumor microenvironment (TME), where they sustain tumor progression and or-tumor immunity. Due to their plasticity, macrophages can exhibit anti- or pro-tumor functions through the expression of different gene sets leading to distinct macrophage phenotypes: M1-like or pro-inflammatory and M2-like or anti-inflammatory. NF-κB transcription factors are central regulators of TAMs in cancers, where they often drive macrophage polarization toward an M2-like phenotype. Therefore, the NF-κB pathway is an attractive therapeutic target for cancer immunotherapy in a wide range of human tumors. Hence, targeting NF-κB pathway in the myeloid compartment is a potential clinical strategy to overcome microenvironment-induced immunosuppression and increase anti-tumor immunity. In this review, we discuss the role of NF-κB as a key driver of macrophage functions in tumors as well as the principal strategies to overcome tumor immunosuppression by targeting the NF-κB pathway.


Asunto(s)
FN-kappa B , Neoplasias , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Macrófagos/metabolismo , Neoplasias/genética , Neoplasias/terapia , Neoplasias/metabolismo , Subunidad p50 de NF-kappa B , Fenotipo , Microambiente Tumoral/genética
7.
Proc Natl Acad Sci U S A ; 107(28): 12611-6, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20616036

RESUMEN

Asbestos carcinogenesis has been linked to the release of cytokines and mutagenic reactive oxygen species (ROS) from inflammatory cells. Asbestos is cytotoxic to human mesothelial cells (HM), which appears counterintuitive for a carcinogen. We show that asbestos-induced HM cell death is a regulated form of necrosis that links to carcinogenesis. Asbestos-exposed HM activate poly(ADP-ribose) polymerase, secrete H(2)O(2), deplete ATP, and translocate high-mobility group box 1 protein (HMGB1) from the nucleus to the cytoplasm, and into the extracellular space. The release of HMGB1 induces macrophages to secrete TNF-alpha, which protects HM from asbestos-induced cell death and triggers a chronic inflammatory response; both favor HM transformation. In both mice and hamsters injected with asbestos, HMGB1 was specifically detected in the nuclei, cytoplasm, and extracellular space of mesothelial and inflammatory cells around asbestos deposits. TNF-alpha was coexpressed in the same areas. HMGB1 levels in asbestos-exposed individuals were significantly higher than in nonexposed controls (P < 0.0001). Our findings identify the release of HMGB1 as a critical initial step in the pathogenesis of asbestos-related disease, and provide mechanistic links between asbestos-induced cell death, chronic inflammation, and carcinogenesis. Chemopreventive approaches aimed at inhibiting the chronic inflammatory response, and especially blocking HMGB1, may decrease the risk of malignant mesothelioma among asbestos-exposed cohorts.


Asunto(s)
Proteína HMGB1/metabolismo , Inflamación/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Adenosina Difosfato Ribosa/farmacología , Animales , Amianto/metabolismo , Amianto/farmacología , Carcinógenos/metabolismo , Carcinógenos/farmacología , Muerte Celular , Núcleo Celular/metabolismo , Células/metabolismo , Cricetinae , Citocinas/metabolismo , Citocinas/farmacología , Células Epiteliales/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Femenino , Proteínas HMGB/metabolismo , Proteínas HMGB/farmacología , Proteína HMGB1/farmacología , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Macrófagos/metabolismo , Mesocricetus , Mesotelioma/metabolismo , Ratones , Ratones Endogámicos BALB C , Necrosis/metabolismo , Neoplasias Pleurales/metabolismo , Poli Adenosina Difosfato Ribosa/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
8.
Proc Natl Acad Sci U S A ; 107(45): 19461-6, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20978210

RESUMEN

CD31 is an Ig-like molecule expressed by leukocytes and endothelial cells with an established role in the regulation of leukocyte trafficking. Despite genetic deletion of CD31 being associated with exacerbation of T cell-mediated autoimmunity, the contribution of this molecule to T-cell responses is largely unknown. Here we report that tumor and allograft rejection are significantly enhanced in CD31-deficient mice, which are also resistant to tolerance induction. We propose that these effects are dependent on an as yet unrecognized role for CD31-mediated homophilic interactions between T cells and antigen-presenting cells (APCs) during priming. We show that loss of CD31 interactions leads to enhanced primary clonal expansion, increased killing capacity, and diminished regulatory functions by T cells. Immunomodulation by CD31 signals correlates with a partial inhibition of proximal T-cell receptor (TCR) signaling, specifically Zap-70 phosphorylation. However, CD31-deficient mice do not develop autoimmunity due to increased T-cell death following activation, and we show that CD31 triggering induces Erk-mediated prosurvival activity in T cells either in conjunction with TCR signaling or autonomously. We conclude that CD31 functions as a nonredundant comodulator of T-cell responses, which specializes in sizing the ensuing immune response by setting the threshold for T-cell activation and tolerance, while preventing memory T-cell death.


Asunto(s)
Tolerancia Inmunológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Linfocitos T/inmunología , Animales , Supervivencia Celular , Células Clonales , Citotoxicidad Inmunológica , Genes de Inmunoglobulinas , Memoria Inmunológica , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética
9.
Arthritis Rheum ; 63(10): 2949-55, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21702006

RESUMEN

OBJECTIVE: Growth arrest and DNA damage-inducible protein 45ß (GADD45ß) is involved in stress responses, cell cycle regulation, and oncogenesis. Previous studies demonstrated that GADD45ß deficiency exacerbates K/BxN serum-induced arthritis and experimental allergic encephalomyelitis (EAE) in mice, indicating that GADD45ß plays a suppressive role in innate and adaptive immune responses. To further understand how GADD45ß regulates autoimmunity, we evaluated collagen-induced arthritis in GADD45ß-/- mice. METHODS: Wild-type (WT) and GADD45ß-/- DBA/1 mice were immunized with bovine type II collagen (CII). Serum anticollagen antibody levels were quantified by enzyme-linked immunosorbent assay. Expression of cytokines and matrix metalloproteinases in the joint and spleen was determined by quantitative polymerase chain reaction. The in vitro T cell cytokine response to CII was measured by multiplex analysis. CD4+CD25+ Treg cells and Th17 cells were quantified using flow cytometry. RESULTS: GADD45ß-/- mice showed significantly lower arthritis severity and joint destruction compared with WT mice. MMP-3 and MMP-13 expression was also markedly reduced in GADD45ß-/- mice. However, serum anti-CII antibody levels were similar in both groups. FoxP3 and interleukin-10 (IL-10) expression was increased 2-3-fold in splenocytes from arthritic GADD45ß-/- mice compared with those from WT mice. Flow cytometric analysis showed greater numbers of CD4+CD25+ Treg cells in the spleen of GADD45ß-/- mice than in the spleen of WT mice. In vitro studies showed that interferon-γ and IL-17 production by T cells was significantly decreased in GADD45ß-/- mice. CONCLUSION: Unlike passive K/BxN arthritis and EAE, GADD45ß deficiency in CIA was associated with lower arthritis severity, elevated IL-10 expression, decreased IL-17 production, and increased numbers of Treg cells. The data suggest that GADD45ß plays a complex role in regulating adaptive immunity and, depending on the model, either enhances or suppresses inflammation.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Artritis Experimental/metabolismo , Inflamación/metabolismo , Articulaciones/metabolismo , Animales , Antígenos de Diferenciación/genética , Artritis Experimental/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Inflamación/genética , Interleucina-10/genética , Interleucina-10/metabolismo , Ratones , Ratones Noqueados , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/metabolismo , Células Th17/metabolismo
10.
Mol Cell Oncol ; 9(1): 2024051, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35252551

RESUMEN

Cancer cells reprogram lipid metabolism to fuel cell division, adaptation to stress, and metastatic dissemination. NF-κB transcription factors control this mechanism in aggressive Consensus Molecular Subtype (CMS)4 of colorectal carcinoma (CRC) via triacylglycerol (TAG) lipase, carboxylesterase 1 (CES1), thereby linking obesity-associated inflammation with metabolic adaptation and cytoprotection from lipid-induced toxicity. Our findings identify a potential therapeutic route to treat patients with metastasis-prone CRC and provide an example for targeting core tumor subtype-based vulnerabilities in cancers beyond CRC.

11.
Biomedicines ; 10(9)2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36140335

RESUMEN

NF-κB transcription factors are major drivers of tumor initiation and progression. NF-κB signaling is constitutively activated by genetic alterations or environmental signals in many human cancers, where it contributes to almost all hallmarks of malignancy, including sustained proliferation, cell death resistance, tumor-promoting inflammation, metabolic reprogramming, tissue invasion, angiogenesis, and metastasis. As such, the NF-κB pathway is an attractive therapeutic target in a broad range of human cancers, as well as in numerous non-malignant diseases. Currently, however, there is no clinically useful NF-κB inhibitor to treat oncological patients, owing to the preclusive, on-target toxicities of systemic NF-κB blockade. In this review, we discuss the principal and most promising strategies being developed to circumvent the inherent limitations of conventional IκB kinase (IKK)/NF-κB-targeting drugs, focusing on new molecules that target upstream regulators or downstream effectors of oncogenic NF-κB signaling, as well as agents targeting individual NF-κB subunits.

12.
J Clin Invest ; 118(5): 1911-23, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18382767

RESUMEN

In the liver, the JNK cascade is induced downstream of TNF receptors (TNFRs) in response to inflammatory, microbial, and toxic challenges. Sustained activation of JNK triggers programmed cell death (PCD), and hepatocyte survival during these challenges requires induction of the NF-kappaB pathway, which antagonizes this activation by upregulating target genes. Thus, modulation of JNK activity is crucial to the liver response to TNFR-mediated challenge. The basis for this modulation, however, is unknown. Here, we investigated the role of the NF-kappaB target Gadd45b in the regulation of hepatocyte fate during liver regeneration after partial hepatectomy. We generated Gadd45b(-/-) mice and found that they exhibited decreased hepatocyte proliferation and increased PCD during liver regeneration. Notably, JNK activity was markedly increased and sustained in livers of Gadd45b(-/-) mice compared with control animals after partial hepatectomy. Furthermore, imposition of a Jnk2-null mutation, attenuating JNK activity, completely rescued the regenerative response in Gadd45b(-/-) mice. Interestingly, Gadd45beta ablation did not affect hepatotoxic JNK signaling after a TNFR-mediated immune challenge, suggesting specificity in the inducible hepatic program for JNK restraint activated during distinct TNFR-mediated challenges. These data provide a basis for JNK suppression during liver regeneration and identify Gadd45beta as a potential therapeutic target in liver diseases.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Supervivencia Celular , Hepatocitos/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Regeneración Hepática/fisiología , Transducción de Señal/fisiología , Animales , Antígenos de Diferenciación/genética , Apoptosis/fisiología , Activación Enzimática , Hepatocitos/citología , Etiquetado Corte-Fin in Situ , Interleucina-6/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Hígado/anatomía & histología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Nat Cell Biol ; 6(2): 146-53, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14743220

RESUMEN

NF-kappa B/Rel transcription factors control apoptosis, also known as programmed cell death. This control is crucial for oncogenesis, cancer chemo-resistance and for antagonizing tumour necrosis factor alpha (TNFalpha)-induced killing. With regard to TNFalpha, the anti-apoptotic activity of NF-kappa B involves suppression of the c-Jun N-terminal kinase (JNK) cascade. Using an unbiased screen, we have previously identified Gadd45 beta/Myd118, a member of the Gadd45 family of inducible factors, as a pivotal mediator of this suppressive activity of NF-kappa B. However, the mechanisms by which Gadd45 beta inhibits JNK signalling are not understood. Here, we identify MKK7/JNKK2--a specific and essential activator of JNK--as a target of Gadd45 beta, and in fact, of NF-kappa B itself. Gadd45 beta binds to MKK7 directly and blocks its catalytic activity, thereby providing a molecular link between the NF-kappa B and JNK pathways. Importantly, Gadd45 beta is required to antagonize TNFalpha-induced cytotoxicity, and peptides disrupting the Gadd45 beta/MKK7 interaction hinder the ability of Gadd45 beta, as well as of NF-kappa B, to suppress this cytotoxicity. These findings establish a basis for the NF-kappa B control of JNK activation and identify MKK7 as a potential target for anti-inflammatory and anti-cancer therapy.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Células Cultivadas , Activación Enzimática , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , MAP Quinasa Quinasa 7 , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Péptidos/genética , Péptidos/metabolismo , Unión Proteica , Alineación de Secuencia , Factor de Necrosis Tumoral alfa/metabolismo
14.
Methods Mol Biol ; 2366: 19-25, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34236630

RESUMEN

Cell fractionation is a method used to study different cellular events like protein translocation and sequestration by disrupting cells and fractionating their contents, thus allowing an enrichment of the protein of interest. Using different concentrations of sucrose or detergent buffer formulations in combination with centrifugations, the cell fractions are separated based on their density and size.


Asunto(s)
Fraccionamiento Celular , FN-kappa B , Fracciones Subcelulares
15.
Methods Mol Biol ; 2366: 27-42, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34236631

RESUMEN

Immunohistochemistry (IHC) is a technique aimed at detecting specific antigens on tissue sections by the use of targeting reagents labeled with reporter molecules. This technique allows a snapshot of the structure of tissue and determines the cellular and subcellular localization of a target antigen. This chapter describes how to identify and localize NF-κB proteins in human tissue using immunohistochemical staining on formalin-fixed paraffin-embedded and frozen tissue.


Asunto(s)
FN-kappa B/metabolismo , Antígenos , Formaldehído , Humanos , Inmunohistoquímica , Adhesión en Parafina , Fosforilación , Fijación del Tejido
16.
Methods Mol Biol ; 2366: 293-303, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34236646

RESUMEN

The reprogramming of cell metabolism is a hallmark of cancer. NF-κB transcription factors coordinate the host defense responses to stress, injury, and infection. They also play a central role in oncogenesis, at least in part by regulating cell metabolism and the adaptation to energy stress conditions in various types of cancer, such as colorectal carcinoma (CRC). Here, we describe the XF Cell Mito Stress Test methodology aimed at characterizing the metabolic and bioenergetic profile of CRC cells following the silencing of the essential NF-κB subunit, RelA. This methodology may also be applied to other cancers to reveal novel core vulnerabilities of malignant cells.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Colorrectales/metabolismo , Metabolismo Energético , Humanos , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Respiración , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
17.
Methods Mol Biol ; 2366: 343-356, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34236649

RESUMEN

Peptides are emerging as an increasingly dependable class of therapeutics in the treatment of cancer and metabolic and cardiovascular diseases, which are all areas of high interest to the pharmaceutical industry. The global market for peptide therapeutics was valued at about 25 billion USD in 2018 and is estimated to reach 57.2 billion USD by the end of 2027. Here, we describe a method for the screening and deconvolution of combinatorial peptide libraries to discover compounds that target discrete signaling components of the NF-κB pathway. Recently, we used this approach to specifically disrupt the interaction between the JNK-activating kinase, MKK7, and the NF-κB-regulated antiapoptotic factor, GADD45ß, in multiple myeloma (MM). We showed that the GADD45ß/MKK7 complex is a functionally critical survival module downstream of NF-κB in MM cells and as such provides an attractive therapeutic target to selectively inhibit NF-κB antiapoptotic signaling in cancer cells. By integrating the library screening and deconvolution methods described here with a rational chemical optimization strategy, we developed the first-in-class GADD45ß/MKK7 inhibitor, DTP3 (a D-tripeptide), which is now being trialed in MM and diffuse large B-cell lymphoma (DLBCL) patients. The same drug discovery approach may be generally applied to therapeutically target other key components of the NF-κB pathway in cancers beyond MM and DLBCL, as well as in non-malignant NF-κB-driven diseases.


Asunto(s)
Transducción de Señal , Apoptosis , Humanos , Linfoma de Células B Grandes Difuso , Mieloma Múltiple/tratamiento farmacológico , FN-kappa B/metabolismo , Biblioteca de Péptidos , Péptidos , Mapeo de Interacción de Proteínas
18.
J Clin Invest ; 131(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33878036

RESUMEN

The ability to adapt to low-nutrient microenvironments is essential for tumor cell survival and progression in solid cancers, such as colorectal carcinoma (CRC). Signaling by the NF-κB transcription factor pathway associates with advanced disease stages and shorter survival in patients with CRC. NF-κB has been shown to drive tumor-promoting inflammation, cancer cell survival, and intestinal epithelial cell (IEC) dedifferentiation in mouse models of CRC. However, whether NF-κB affects the metabolic adaptations that fuel aggressive disease in patients with CRC is unknown. Here, we identified carboxylesterase 1 (CES1) as an essential NF-κB-regulated lipase linking obesity-associated inflammation with fat metabolism and adaptation to energy stress in aggressive CRC. CES1 promoted CRC cell survival via cell-autonomous mechanisms that fuel fatty acid oxidation (FAO) and prevent the toxic build-up of triacylglycerols. We found that elevated CES1 expression correlated with worse outcomes in overweight patients with CRC. Accordingly, NF-κB drove CES1 expression in CRC consensus molecular subtype 4 (CMS4), which is associated with obesity, stemness, and inflammation. CES1 was also upregulated by gene amplifications of its transcriptional regulator HNF4A in CMS2 tumors, reinforcing its clinical relevance as a driver of CRC. This subtype-based distribution and unfavorable prognostic correlation distinguished CES1 from other intracellular triacylglycerol lipases and suggest CES1 could provide a route to treat aggressive CRC.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Neoplasias Colorrectales/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Triglicéridos/metabolismo , Hidrolasas de Éster Carboxílico/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Proteínas de Neoplasias/genética , Triglicéridos/genética
19.
Dev Cell ; 8(4): 452-4, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15809025

RESUMEN

Proinflammatory cytokine TNFalpha triggers cell death by inducing reactive oxygen species (ROS). These then inflict cytotoxicity through downstream activation of the JNK MAPK cascade. Yet the mechanisms by which ROS trigger JNK signaling have remained elusive. In a recent issue of Cell, Kamata et al. now provide one such mechanism.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Oxígeno/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/fisiología , Activación Enzimática , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Gastroenterology ; 136(2): 694-704.e4, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18952083

RESUMEN

BACKGROUND & AIMS: The ability of the liver to regenerate hepatic mass is essential to withstanding liver injury. The process of liver regeneration is tightly regulated by distinct signaling cascades involving components of the innate immune system, cytokines, and growth factors. However, the role of the adaptive immune system in regulation of liver regeneration is not well-defined. The role of adaptive immune system in liver regeneration was investigated in lymphocyte-deficient mice and in conditional lymphotoxin-deficient mice. METHODS: A model of liver regeneration after 70% partial hepatectomy was used, followed by examination of liver pathology, survival, DNA synthesis, and cytokine expression. RESULTS: We found that mice deficient in T cells show a reduced capacity for liver regeneration following partial hepatectomy. Furthermore, surface lymphotoxin, provided by T cells, is critical for liver regeneration. Mice specifically deficient in T-cell lymphotoxin had increased liver damage and a reduced capacity to initiate DNA synthesis after partial hepatectomy. Transfer of splenocytes from wild-type but not lymphotoxin-deficient mice improved liver regeneration in T cell-deficient mice. We found that an agonistic antibody against the lymphotoxin beta receptor was able to facilitate liver regeneration by reducing liver injury, increasing interleukin-6 production, hepatocyte DNA synthesis, and survival of lymphocyte-deficient (Rag) mice after partial hepatectomy. CONCLUSIONS: The adaptive immune system directly regulates liver regeneration via a T cell-derived lymphotoxin axis, and pharmacological stimulation of lymphotoxin beta receptor might represent a novel therapeutic approach to improve liver regeneration.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Regeneración Hepática/fisiología , Linfotoxina-alfa/metabolismo , Linfotoxina beta/metabolismo , Animales , Hepatectomía , Sistema Inmunológico/fisiología , Interleucina-6/metabolismo , Hígado/citología , Hígado/metabolismo , Hígado/cirugía , Receptor beta de Linfotoxina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Bazo/citología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda