Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(3): e1011241, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36930690

RESUMEN

Dengue virus (DENV) infection can induce life-threatening dengue hemorrhagic fever/dengue shock syndrome in infected patients. DENV is a threat to global health due to its growing numbers and incidence of infection in the last 50 years. During infection, DENV expresses ten structural and nonstructural proteins modulating cell responses to benefit viral replication. However, the lack of knowledge regarding the cellular proteins and their functions in enhancing DENV pathogenesis impedes the development of antiviral drugs and therapies against fatal DENV infection. Here, we identified that integrin-linked kinase (ILK) is a novel enhancing factor for DENV infection by suppressing type I interferon (IFN) responses. Mechanistically, ILK binds DENV NS1 and NS3, activates Akt and Erk, and induces NF-κB-driven suppressor of cytokine signaling 3 (SOCS3) expression. Elevated SOCS3 in DENV-infected cells inhibits phosphorylation of STAT1/2 and expression of interferon-stimulated genes (ISGs). Inhibiting ILK, Akt, or Erk activation abrogates SOCS3 expression. In DENV-infected mice, the treatment of an ILK inhibitor significantly reduces viral loads in the brains, disease severity, and mortality rate. Collectively, our results show that ILK is a potential therapeutic target against DENV infection.


Asunto(s)
Virus del Dengue , Dengue , Interferón Tipo I , Animales , Ratones , Virus del Dengue/fisiología , Proteínas Proto-Oncogénicas c-akt , Replicación Viral , Interferón Tipo I/uso terapéutico
2.
PLoS Pathog ; 18(4): e1010469, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35486576

RESUMEN

Dengue virus (DENV) which infects about 390 million people per year in tropical and subtropical areas manifests various disease symptoms, ranging from fever to life-threatening hemorrhage and even shock. To date, there is still no effective treatment for DENV disease, but only supportive care. DENV nonstructural protein 1 (NS1) has been shown to play a key role in disease pathogenesis. Recent studies have shown that anti-DENV NS1 antibody can provide disease protection by blocking the DENV-induced disruption of endothelial integrity. We previously demonstrated that anti-NS1 monoclonal antibody (mAb) protected mice from all four serotypes of DENV challenge. Here, we generated humanized anti-NS1 mAbs and transferred them to mice after DENV infection. The results showed that DENV-induced prolonged bleeding time and skin hemorrhage were reduced, even several days after DENV challenge. Mechanistic studies showed the ability of humanized anti-NS1 mAbs to inhibit NS1-induced vascular hyperpermeability and to elicit Fcγ-dependent complement-mediated cytolysis as well as antibody-dependent cellular cytotoxicity of cells infected with four serotypes of DENV. These results highlight humanized anti-NS1 mAb as a potential therapeutic agent in DENV infection.


Asunto(s)
Virus del Dengue , Dengue , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Dengue/prevención & control , Modelos Animales de Enfermedad , Hemorragia/etiología , Humanos , Ratones , Proteínas no Estructurales Virales/metabolismo
3.
J Biomed Sci ; 31(1): 10, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38243273

RESUMEN

BACKGROUND: The tumor microenvironment is characterized by inflammation-like and immunosuppression situations. Although cancer-associated fibroblasts (CAFs) are among the major stromal cell types in various solid cancers, including colon cancer, the interactions between CAFs and immune cells remains largely uncharacterized. Pentraxin 3 (PTX3) is responsive to proinflammatory cytokines and modulates immunity and tissue remodeling, but its involvement in tumor progression appears to be context-dependent and is unclear. METHODS: Open-access databases were utilized to examine the association of PTX3 expression and the fibroblast signature in colon cancer. Loss-of-function assays, including studies in tamoxifen-induced Ptx3 knockout mice and treatment with an anti-PTX3 neutralizing antibody (WHC-001), were conducted to assess the involvement of PTX3 in colon cancer progression as well as its immunosuppressive effect. Finally, bioinformatic analyses and in vitro assays were performed to reveal the downstream effectors and decipher the involvement of the CREB1/CEBPB axis in response to PTX3 and PTX3-induced promotion of M2 macrophage polarization. RESULTS: Clinically, higher PTX3 expression was positively correlated with fibroblasts and inflammatory response signatures and associated with a poor survival outcome in colon cancer patients. Blockade of PTX3 significantly reduced stromal cell-mediated tumor development. The decrease of the M2 macrophage population and an increase of the cytotoxic CD8+ T-cell population were observed following PTX3 inactivation in allografted colon tumors. We further revealed that activation of cyclic AMP-responsive element-binding protein 1 (CREB1) mediated the PTX3-induced promotion of M2 macrophage polarization. CONCLUSIONS: PTX3 contributes to stromal cell-mediated protumor immunity by increasing M2-like macrophage polarization, and inhibition of PTX3 with WHC-001 is a potential therapeutic strategy for colon cancer.


Asunto(s)
Neoplasias del Colon , Macrófagos , Componente Amiloide P Sérico , Animales , Ratones , Humanos , Macrófagos/metabolismo , Proteína C-Reactiva/genética , Neoplasias del Colon/genética , Terapia de Inmunosupresión , Microambiente Tumoral
4.
IUBMB Life ; 74(2): 170-179, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34553486

RESUMEN

Autophagy is not only an intracellular recycling degradation system that maintains cellular homeostasis but is also a component of innate immunity that contributes to host defense against viral infection. The viral components as well as viral particles trapped in autophagosomes can be delivered to lysosomes for degradation. Abundant evidence indicates that dengue virus (DENV) has evolved the potent ability to hijack or subvert autophagy process for escaping host immunity and promoting viral replication. Moreover, autophagy is often required to deliver viral components to pattern recognition receptors signaling for interferon (IFN)-mediated viral elimination. Hence, this review summarizes DENV-induced autophagy, which exhibits dual effects on proviral activity of promoting replication and antiviral activity to eliminating viral particles.


Asunto(s)
Virus del Dengue , Dengue , Virosis , Autofagia , Dengue/genética , Humanos , Inmunidad Innata , Transducción de Señal , Replicación Viral
5.
IUBMB Life ; 74(2): 157-169, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34467634

RESUMEN

Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are key cells in regulating tumor development, metastasis, immune responses, inflammation, and chemoresistance. In response to TME stimulation, circulating monocytes are recruited and differentiated as TAMs. Most TAMs are defined as alternatively activated (M2) phenotype to create immunosuppressive TME and support tumor progression. In contrast, classically activated (M1) TAMs can produce pro-inflammatory cytokines and enhance immune responses against tumor development. Autophagy is a conserved catabolic process to control cellular homeostasis and biological function. Emerging evidence reveals crucial contribution of autophagy in modulating TAM plasticity and functional polarization in TME. In this review, we introduce the current understanding of autophagy-regulated TAM function in development of cancer. We focus on how autophagy modulates antigen presentation, LC3-associated phagocytosis, cytokine secretion, inflammasome regulation, recruitment, differentiation, and polarization of TAMs and suggest strategies for potential therapeutics by targeting autophagy in TAMs. We expect this review can provide a new notion of future cancer immunotherapy.


Asunto(s)
Neoplasias , Macrófagos Asociados a Tumores , Autofagia , Humanos , Inmunoterapia , Neoplasias/metabolismo , Microambiente Tumoral
6.
J Biomed Sci ; 29(1): 56, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35927755

RESUMEN

All cells in the changing tumor microenvironment (TME) need a class of checkpoints to regulate the balance among exocytosis, endocytosis, recycling and degradation. The vesicular trafficking and secretion pathways regulated by the small Rab GTPases and their effectors convey cell growth and migration signals and function as meditators of intercellular communication and molecular transfer. Recent advances suggest that Rab proteins govern conventional and unconventional vesicular secretion pathways by trafficking widely diverse cargoes and substrates in remodeling TME. The mechanisms underlying the regulation of conventional and unconventional vesicular secretion pathways, their action modes and impacts on the cancer and stromal cells have been the focus of much attention for the past two decades. In this review, we discuss the current understanding of vesicular secretion pathways in TME. We begin with an overview of the structure, regulation, substrate recognition and subcellular localization of vesicular secretion pathways. We then systematically discuss how the three fundamental vesicular secretion processes respond to extracellular cues in TME. These processes are the conventional protein secretion via the endoplasmic reticulum-Golgi apparatus route and two types of unconventional protein secretion via extracellular vesicles and secretory autophagy. The latest advances and future directions in vesicular secretion-involved interplays between tumor cells, stromal cell and host immunity are also described.


Asunto(s)
Vías Secretoras , Microambiente Tumoral , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Transporte de Proteínas , Proteínas de Unión al GTP rab/metabolismo
7.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34638904

RESUMEN

Group A Streptococcus (GAS) causes invasive human diseases with the cytokine storm. Interleukin-33 (IL-33)/suppression of tumorigenicity 2 (ST2) axis is known to drive TH2 response, while its effect on GAS infection is unclear. We used an air pouch model to examine the effect of the IL-33/ST2 axis on GAS-induced necrotizing fasciitis. GAS infection induced IL-33 expression in wild-type (WT) C57BL/6 mice, whereas the IL-33- and ST2-knockout mice had higher mortality rates, more severe skin lesions and higher bacterial loads in the air pouches than those of WT mice after infection. Surveys of infiltrating cells in the air pouch of GAS-infected mice at the early stage found that the number and cell viability of infiltrating cells in both gene knockout mice were lower than those of WT mice. The predominant effector cells in GAS-infected air pouches were neutrophils. Absence of the IL-33/ST2 axis enhanced the expression of inflammatory cytokines, but not TH1 or TH2 cytokines, in the air pouch after infection. Using in vitro assays, we found that the IL-33/ST2 axis not only enhanced neutrophil migration but also strengthened the bactericidal activity of both sera and neutrophils. These results suggest that the IL-33/ST2 axis provided the protective effect on GAS infection through enhancing the innate immunity.


Asunto(s)
Inmunidad Innata/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Movimiento Celular/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/genética , Interleucina-33/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/microbiología , Transducción de Señal/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/fisiología
8.
Adv Exp Med Biol ; 1240: 83-93, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32060890

RESUMEN

Suppression of tumorigenicity 2 (ST2), also known as interleukin-1 receptor-like 1 (IL1RL1), is one of the natural receptors of IL-33. Three major isoforms, ST2L (transmembrane form), sST2 (soluble form), and ST2V, are generated by alternative splicing. Damage to stromal cells induces necrosis and release of IL-33, which binds to heterodimeric ST2L/IL-1RAcP complex on the membrane of a variety of immune cells. This IL-33/ST2L signal induces transcription of the downstream inflammatory and anti-inflammatory genes by activating diverse intracellular kinases and factors to mount an adequate immune response, even in tumor microenvironment. For example, activation of IL-33/ST2L signal may trigger Th2-dependent M2 macrophage polarization to facilitate tumor progression. Notably, sST2 is a soluble form of ST2 that lacks a transmembrane domain but preserves an extracellular domain similar to ST2L, which acts as a "decoy" receptor for IL-33. sST2 has been shown to involve in the inflammatory tumor microenvironment and the progression of colorectal cancer, non-small cell lung cancer, and gastric cancer. Therefore, targeting the IL-33/ST2 axis becomes a promising new immunotherapy for treatment of many cancers. This chapter reviews the recent findings on IL-33/ST2L signaling in tumor microenvironment, the trafficking mode of sST2, and the pharmacological strategies to target IL-33/ST2 axis for cancer treatment.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Transducción de Señal , Microambiente Tumoral , Animales , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/antagonistas & inhibidores , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/antagonistas & inhibidores , Interleucina-33/inmunología , Interleucina-33/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología
9.
Int J Mol Sci ; 21(24)2020 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-33352639

RESUMEN

Dengue virus (DENV) infection is a significant public health threat in tropical and subtropical regions; however, there is no specific antiviral drug. Accumulated studies have revealed that DENV infection induces several cellular responses, including autophagy and apoptosis. The crosstalk between autophagy and apoptosis is associated with the interactions among components of these two pathways, such as apoptotic caspase-mediated cleavage of autophagy-related proteins. Here, we show that DENV-induced autophagy inhibits early cell apoptosis and hence enhances DENV replication. Later, the apoptotic activities are elevated to suppress autophagy through cleavage of Beclin-1, an essential autophagy-related protein. Inhibition of cleavage of Beclin-1 by a pan-caspase inhibitor, Z-VAD, increases both autophagy and viral replication. Regarding the mechanism, we further found that DENV nonstructural protein 1 (NS1) is able to interact with Beclin-1 during DENV infection. The interaction between Beclin-1 and NS1 attenuates Beclin-1 cleavage and facilitates autophagy to prevent cell apoptosis. Our study suggests a novel mechanism whereby NS1 preserves Beclin-1 for maintaining autophagy to antagonize early cell apoptosis; however, elevated caspases trigger apoptosis by degrading Beclin-1 in the late stage of infection. These findings suggest implications for anti-DENV drug design.


Asunto(s)
Beclina-1/metabolismo , Caspasas/metabolismo , Virus del Dengue/aislamiento & purificación , Dengue/patología , Dengue/virología , Proteínas no Estructurales Virales/metabolismo , Células A549 , Aedes , Animales , Autofagia , Dengue/metabolismo , Humanos
10.
J Immunol ; 199(8): 2834-2844, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28904127

RESUMEN

Dengue virus (DENV) is the causative agent of dengue fever, dengue hemorrhagic fever, and dengue shock syndrome and is endemic to tropical and subtropical regions of the world. Our previous studies showed the existence of epitopes in the C-terminal region of DENV nonstructural protein 1 (NS1) which are cross-reactive with host Ags and trigger anti-DENV NS1 Ab-mediated endothelial cell damage and platelet dysfunction. To circumvent these potentially harmful events, we replaced the C-terminal region of DENV NS1 with the corresponding region from Japanese encephalitis virus NS1 to create chimeric DJ NS1 protein. Passive immunization of DENV-infected mice with polyclonal anti-DJ NS1 Abs reduced viral Ag expression at skin inoculation sites and shortened DENV-induced prolonged bleeding time. We also investigated the therapeutic effects of anti-NS1 mAb. One mAb designated 2E8 does not recognize the C-terminal region of DENV NS1 in which host-cross-reactive epitopes reside. Moreover, mAb 2E8 recognizes NS1 of all four DENV serotypes. We also found that mAb 2E8 caused complement-mediated lysis in DENV-infected cells. In mouse model studies, treatment with mAb 2E8 shortened DENV-induced prolonged bleeding time and reduced viral Ag expression in the skin. Importantly, mAb 2E8 provided therapeutic effects against all four serotypes of DENV. We further found that mAb administration to mice as late as 1 d prior to severe bleeding still reduced prolonged bleeding time and hemorrhage. Therefore, administration with a single dose of mAb 2E8 can protect mice against DENV infection and pathological effects, suggesting that NS1-specific mAb may be a therapeutic option against dengue disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Virus del Dengue/inmunología , Dengue/terapia , Hemorragia/prevención & control , Inmunoterapia/métodos , Proteínas no Estructurales Virales/metabolismo , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Autoantígenos/inmunología , Células Cultivadas , Reacciones Cruzadas , Dengue/complicaciones , Dengue/inmunología , Virus del Dengue/genética , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/genética , Epítopos/genética , Hemorragia/etiología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Proteínas Recombinantes/inmunología , Factor de Transcripción STAT1/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
11.
Int J Cancer ; 143(7): 1753-1763, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717487

RESUMEN

Interplay between cancer epithelial cells and the surrounding immune cells shape the tumor microenvironment to promote cancer progression. Tumor-associated macrophages are well recognized for their roles in cancer progression. Accumulating evidence also indicates implication of Rab small GTPase-mediated exocytosis in tumorigenesis. However, the mechanism for Rab-mediated exocytosis in regulation of macrophage polarization is not clear. We have previously identified Rab37 as a metastasis suppressor in lung cancer. In our study, we identified a novel Rab37 trafficking cargo soluble ST2 (sST2), which skewed macrophage polarization toward anti-tumoral M1-like phenotype in vitro. We further demonstrated that Rab37-mediated sST2 secretion significantly increased the ratio of M1 vs. M2 in xenografts and thus reduced tumor growth. Moreover, lung cancer patients with low Rab37, low sST2 and low ratio of M1 vs. M2 macrophages expression profile correlated with worse overall survival examined by Kaplan-Meier survival analysis. Multivariate Cox regression analysis showed that this Rab37-sST2-M1/M2 expression profile predicted poor prognosis. Our findings reveal a novel regulation of cancerous Rab37 in microenvironmental macrophages polarization, which preferentially shifts to anti-tumoral phenotype and thereby suppresses lung tumor growth.


Asunto(s)
Exocitosis , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Neoplasias Pulmonares/patología , Macrófagos/patología , Proteínas de Unión al GTP rab/metabolismo , Animales , Apoptosis , Movimiento Celular , Proliferación Celular , Humanos , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fenotipo , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Artículo en Inglés | MEDLINE | ID: mdl-29581121

RESUMEN

Group A Streptococcus (GAS) is an important human pathogen that causes a wide spectrum of diseases, including necrotizing fasciitis and streptococcal toxic shock syndrome. Dextromethorphan (DM), an antitussive drug, has been demonstrated to efficiently reduce inflammatory responses, thereby contributing to an increased survival rate of GAS-infected mice. However, the anti-inflammatory mechanisms underlying DM treatment in GAS infection remain unclear. DM is known to exert neuroprotective effects through an NADPH oxidase-dependent regulated process. In the present study, membrane translocation of NADPH oxidase subunit p47phox and subsequent reactive oxygen species (ROS) generation induced by GAS infection were significantly inhibited via DM treatment in RAW264.7 murine macrophage cells. Further determination of proinflammatory mediators revealed that DM effectively suppressed inducible nitric oxide synthase (iNOS) expression and NO, tumor necrosis factor alpha, and interleukin-6 generation in GAS-infected RAW264.7 cells as well as in air-pouch-infiltrating cells from GAS/DM-treated mice. GAS infection caused AKT dephosphorylation, glycogen synthase kinase-3ß (GSK-3ß) activation, and subsequent NF-κB nuclear translocation, which were also markedly inhibited by treatment with DM and an NADPH oxidase inhibitor, diphenylene iodonium. These results suggest that DM attenuates GAS infection-induced overactive inflammation by inhibiting NADPH oxidase-mediated ROS production that leads to downregulation of the GSK-3ß/NF-κB/NO signaling pathway.


Asunto(s)
Dextrometorfano/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/metabolismo , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/enzimología , Animales , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Compuestos Onio/farmacología , Oxidación-Reducción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Infecciones Estreptocócicas/metabolismo , Células THP-1
13.
Nanomedicine ; 10(4): 819-29, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24333595

RESUMEN

Magnetic manganese ferrite (MnFe2O4) nanoparticles with approximately 100nm in diameter were used to improve the performance of an immunoassay for detecting influenza infections. The synthesized nanoparticles were tested for long-term storage to confirm the stability of their thermal decomposition process. Then, an integrated microfluidic system was developed to perform the diagnosis process automatically, including virus purification and detection. To apply these nanoparticles for influenza diagnosis, a micromixer was optimized to reduce the dead volume within the microfluidic chip. Furthermore, the mixing index of the micromixer could achieve as high as 97% in 2seconds. The optical signals showed that this nanoparticle-based immunoassay with dynamic mixing could successfully achieve a detection limit of influenza as low as 0.007 HAU. When compared with the 4.5-µm magnetic beads, the optical signals of the MnFe2O4 nanoparticles were twice as sensitive. Furthermore, five clinical specimens were tested to verify the usability of the developed system. FROM THE CLINICAL EDITOR: In this study, magnetic manganese ferrite nanoparticles were used to improve the performance of a novel immunoassay for the rapid and efficient detection of influenza infections.


Asunto(s)
Óxido Ferrosoférrico/química , Gripe Humana/diagnóstico , Técnicas Analíticas Microfluídicas/métodos , Nanopartículas/química , Orthomyxoviridae/inmunología , Animales , Perros , Humanos , Inmunoensayo/métodos , Células de Riñón Canino Madin Darby , Técnicas Analíticas Microfluídicas/instrumentación , Orthomyxoviridae/química , Sensibilidad y Especificidad
14.
Cell Death Dis ; 15(5): 356, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778059

RESUMEN

IL-33 is a danger signal that binds to its receptor ST2L to promote tumor progression. This study identifies the IL-33/ST2L positive-feedback loop and the trafficking of ST2L membrane presentation in macrophages that contribute to lung tumor progression. Mechanistically, IL-33 induces ST2L upregulation by activating NF-κB, which binds to the promoter region of the ST2L gene. Moreover, Rab37, a small GTPase involved in membrane trafficking, mediates ST2L trafficking to the plasma membrane of M2 macrophages. This IL-33/NF-κB/ST2L/Rab37 axis promotes positive-feedback loops that enhance ST2L expression and membrane trafficking in M2 macrophages. Notably, neutralizing antibodies against IL-33 or ST2L block NF-κB activity, suppress M2 macrophage polarization, and synergistically inhibit tumor growth when combined with cisplatin treatment in vitro/vivo. Clinically, Rab37+/ST2L+/CD206+ tumor-infiltrating M2 macrophages correlate with advanced-stage lung cancer patients with poor response to chemotherapy. These findings unveil a positive-feedback mechanism and provide a basis for IL-33/ST2L-targeting therapy for cancer.


Asunto(s)
Interleucina-33 , Neoplasias Pulmonares , Macrófagos , FN-kappa B , Proteínas de Unión al GTP rab , Interleucina-33/metabolismo , Interleucina-33/genética , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , FN-kappa B/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Animales , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab/genética , Ratones , Retroalimentación Fisiológica , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Ratones Endogámicos C57BL , Femenino
15.
Biomed Pharmacother ; 176: 116825, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38820971

RESUMEN

Considering the limited efficacy of current therapies in lung, colorectal, and pancreatic cancers, innovative combination treatments with diverse mechanisms of action are needed to improve patients' outcomes. Chitinase-3 like-1 protein (CHI3L1) emerges as a versatile factor with significant implications in various diseases, particularly cancers, fostering an immunosuppressive tumor microenvironment for cancer progression. Therefore, pre-clinical validation is imperative to fully realize its potential in cancer treatment. We developed phage display-derived fully human monoclonal CHI3L1 neutralizing antibodies (nAbs) and verified the nAbs-antigen binding affinity and specificity in lung, pancreatic and colorectal cancer cell lines. Tumor growth signals, proliferation and migration ability were all reduced by CHI3L1 nAbs in vitro. Orthotopic or subcutaneous tumor mice model and humanized mouse model were established for characterizing the anti-tumor properties of two CHI3L1 nAb leads. Importantly, CHI3L1 nAbs not only inhibited tumor growth but also mitigated fibrosis, angiogenesis, and restored immunostimulatory functions of immune cells in pancreatic, lung, and colorectal tumor mice models. Mechanistically, CHI3L1 nAbs directly suppressed the activation of pancreatic stellate cells and the transformation of macrophages into myofibroblasts, thereby attenuating fibrosis. These findings strongly support the therapeutic potential of CHI3L1 nAbs in overcoming clinical challenges, including the failure of gemcitabine in pancreatic cancer.


Asunto(s)
Anticuerpos Monoclonales , Proliferación Celular , Proteína 1 Similar a Quitinasa-3 , Neoplasias Colorrectales , Fibrosis , Neoplasias Pulmonares , Neovascularización Patológica , Neoplasias Pancreáticas , Animales , Proteína 1 Similar a Quitinasa-3/metabolismo , Proteína 1 Similar a Quitinasa-3/antagonistas & inhibidores , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Ratones , Línea Celular Tumoral , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Anticuerpos Neutralizantes/farmacología , Antineoplásicos Inmunológicos/farmacología , Angiogénesis
16.
Virus Res ; 336: 199203, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37625648

RESUMEN

Fulminant hepatitis is a life-threatening complication of coxsackievirus B (CVB) 3 infections. The condition may deteriorate to disseminated intravascular coagulopathy with markedly increased liver enzymes, inflammatory cytokines, and chemokines, which significantly induce local and systemic inflammation. Curcumin exhibits anti-inflammatory and antiviral characteristics in inflammatory and infectious diseases. Here we determined effects of curcumin on viral replications, cytokine and chemokine expressions, and liver damage in CVB3-infected Huh-7 cells. The mouse-adapted CVB3 strain was used to investigate the antiviral and anti-inflammatory effects of curcumin on CVB3-induced hepatitis in a mouse model. In vitro studies showed that curcumin reduced viral protein and titer levels and increased cell viability. Curcumin enhanced the heme oxygenase-1 (HO-1) protein level and decreased the levels of cleaved caspase-3 protein and mRNA of gene encoding C-X-C motif chemokine 10 in infected cells. In vivo studies showed that curcumin improved the survival rate and clinical scores in mice and reduced the viral titer in the liver during CVB3 infection. Moreover, the HO-1 levels were increased, and the cleaved caspase-3 levels were diminished in the CVB3-infected liver. Curcumin reduced the levels of interferon (IFN)-γ and monokine induced by IFN-γ in liver and levels of interleukin (IL)-8 in serum, but increased levels of regulated activation, normal T cell expression in liver and levels of IL-10 in serum of CVB3-infected mice. In summary, curcumin presents antiviral and anti-inflammation efficacies in CVB3 infection in vitro and in vivo; these results provide potential evidence on the feasibility of curcumin for clinical treatment.

17.
J Exp Clin Cancer Res ; 42(1): 22, 2023 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-36639681

RESUMEN

BACKGROUND: Nitric oxide-releasing drugs are used for cardiovascular diseases; however, their effects on the tumor immune microenvironment are less clear. Therefore, this study explored the impact of nitric oxide donors on tumor progression in immune-competent mice. METHODS: The effects of three different nitric oxide-releasing compounds (SNAP, SNP, and ISMN) on tumor growth were studied in tumor-bearing mouse models. Three mouse tumor models were used: B16F1 melanoma and LL2 lung carcinoma in C57BL/6 mice, CT26 colon cancer in BALB/c mice, and LL2 lung carcinoma in NOD/SCID mice. After nitric oxide treatment, splenic cytokines and lymphocytes were analyzed by cytokine array and flow cytometry, and tumor-infiltrating lymphocytes in the TME were analyzed using flow cytometry and single-cell RNA sequencing. RESULTS: Low doses of three exogenous nitric oxide donors inhibited tumor growth in two immunocompetent mouse models but not in NOD/SCID immunodeficient mice. Low-dose nitric oxide donors increase the levels of splenic cytokines IFN-γ and TNF-α but decrease the levels of cytokines IL-6 and IL-10, suggesting an alteration in Th2 cells. Nitric oxide donors increased the number of CD8+ T cells with activation gene signatures, as indicated by single-cell RNA sequencing. Flow cytometry analysis confirmed an increase in infiltrating CD8+ T cells and dendritic cells. The antitumor effect of nitric oxide donors was abolished by depletion of CD8+ T cells, indicating the requirement for CD8+ T cells. Tumor inhibition correlated with a decrease in a subtype of protumor macrophages and an increase in a subset of Arg1-positive macrophages expressing antitumor gene signatures. The increase in this subset of macrophages was confirmed by flow cytometry analysis. Finally, the combination of low-dose nitric oxide donor and cisplatin induced an additive cancer therapeutic effect in two immunocompetent animal models. The enhanced therapeutic effect was accompanied by an increase in the cells expressing the gene signature of NK cell. CONCLUSIONS: Low concentrations of exogenous nitric oxide donors inhibit tumor growth in vivo by regulating T cells and macrophages. CD8+ T cells are essential for antitumor effects. In addition, low-dose nitric oxide donors may be combined with chemotherapeutic drugs in cancer therapy in the future.


Asunto(s)
Linfocitos T CD8-positivos , Carcinoma , Animales , Ratones , Óxido Nítrico , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Reposicionamiento de Medicamentos , Ratones Endogámicos C57BL , Ratones SCID , Citocinas , Microambiente Tumoral
18.
Immunology ; 136(2): 139-52, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22121944

RESUMEN

MHC class I-restricted CD8 T-lymphocyte epitopes comprise anchor motifs, T-cell receptor (TCR) contact residues and the peptide backbone. Serial variant epitopes with substitution of amino acids at either anchor motifs or TCR contact residues have been synthesized for specific interferon-γ responses to clarify the TCR recognition mechanism as well as to assess the epitope prediction capacity of immunoinformatical programmes. CD8 T lymphocytes recognise the steric configuration of functional groups at the TCR contact side chain with a parallel observation that peptide backbones of various epitopes adapt to the conserved conformation upon binding to the same MHC class I molecule. Variant epitopes with amino acid substitutions at the TCR contact site are not recognised by specific CD8 T lymphocytes without compromising their binding capacity to MHC class I molecules, which demonstrates two discrete antigen presentation events for the binding of peptides to MHC class I molecules and for TCR recognition. The predicted outcome of immunoinformatical programmes is not consistent with the results of epitope identification by laboratory experiments in the absence of information on the interaction with TCR contact residues. Immunoinformatical programmes based on the binding affinity to MHC class I molecules are not sufficient for the accurate prediction of CD8 T-lymphocyte epitopes. The predictive capacity is further improved to distinguish mutant epitopes from the non-mutated epitopes if the peptide-TCR interface is integrated into the computing simulation programme.


Asunto(s)
Mapeo Epitopo , Epítopos Inmunodominantes/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Biología Computacional , Genes MHC Clase I/inmunología , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Mutación , Conformación Proteica , Receptores de Antígenos de Linfocitos T/química , Infecciones por Virus Sincitial Respiratorio/inmunología , Análisis de Secuencia de Proteína , Programas Informáticos
19.
Front Cell Infect Microbiol ; 12: 814307, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35350437

RESUMEN

Histone modifications control the lytic gene expression of herpes simplex virus 1 (HSV-1). The heterochromatin mark, trimethylation of histone H3 on lysine (K) 9 (H3K9me3), is detected on HSV-1 genomes at early phases of infection to repress viral gene transcription. However, the components and mechanisms involved in the process are mostly unknown. Integrin-linked kinase (ILK) is activated by PI3K to phosphorylate Akt and promote several RNA virus infections. Akt has been shown to enhance HSV-1 infection, suggesting a pro-viral role of ILK in HSV-1 infection that has not been addressed before. Here, we reveal that ILK enhances HSV-1 replication in an Akt-independent manner. ILK reduces the accumulation of H3K9me3 on viral promoters and replication compartments. Notably, ILK reduces H3K9me3 in a manner independent of ICP0. Instead, we show an increased binding of H3K9 methyltransferase SUV39H1 and corepressor TRIM28 on viral promoters in ILK knockdown cells. Knocking down SUV39H1 or TRIM28 increases HSV-1 lytic gene transcription in ILK knockdown cells. These results show that ILK antagonizes SVU39H1- and TRIM28-mediated repression on lytic gene transcription. We further demonstrate that ILK knockdown reduces TRIM28 phosphorylation on serine 473 and 824 in HSV-1-infected cells, suggesting that ILK facilitates TRIM28 phosphorylation to abrogate its inhibition on lytic gene transcription. OSU-T315, an ILK inhibitor, suppresses HSV-1 replication in cells and mice. In conclusion, we demonstrate that ILK decreases H3K9me3 on HSV-1 DNA by reducing SUV39H1 and TRIM28 binding. Moreover, our results suggest that targeting ILK could be a broad-spectrum antiviral strategy for DNA and RNA virus infections, especially for DNA viruses controlled by histone modifications.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Animales , Herpes Simple/metabolismo , Herpesvirus Humano 1/genética , Histonas/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/genética
20.
Theranostics ; 12(1): 340-361, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34987649

RESUMEN

Background: Chitinase 3-like-1 (CHI3L1) is a secretion glycoprotein associated with the immunosuppressive tumor microenvironment (TME). The secretory mode of CHI3L1 makes it a promising target for cancer treatment. We have previously reported that Rab37 small GTPase mediates secretion of IL-6 in macrophages to promote cancer progression, whereas the roles of Rab37 in the intracellular trafficking and exocytosis of CHI3L1 are unclear. Methods: We examined the concentration of CHI3L1 in the culture medium of splenocytes and bone marrow derived macrophages (BMDMs) from wild-type or Rab37 knockout mice, and macrophage or T cell lines expressing wild type, active GTP-bound or inactive GDP-bound Rab37. Vesicle isolation, total internal reflection fluorescence microscopy, and real-time confocal microscopy were conducted. We developed polyclonal neutralizing-CHI3L1 antibodies (nCHI3L1 Abs) to validate the therapeutic efficacy in orthotopic lung, pancreas and colon cancer allograft models. Multiplex fluorescence immunohistochemistry was performed to detect the protein level of Rab37 and CHI3L1, and localization of the tumor-infiltrating immune cells in allografts from mice or tumor specimens from cancer patients. Results: We demonstrate a novel secretion mode of CHI3L1 mediated by the small GTPase Rab37 in T cells and macrophages. Rab37 mediated CHI3L1 intracellular vesicle trafficking and exocytosis in a GTP-dependent manner, which is abolished in the splenocytes and BMDMs from Rab37 knockout mice and attenuated in macrophage or T cell lines expressing the inactive Rab37. The secreted CHI3L1 activated AKT, ß-catenin and NF-κB signal pathways in cancer cells and macrophages to foster a protumor TME characterized by activating M2 macrophages and increasing the population of regulatory T cells. Our developed nCHI3L1 Abs showed the dual properties of reducing tumor growth/metastases and eliciting an immunostimulatory TME in syngeneic orthotopic lung, pancreas and colon tumor models. Clinically, high plasma level or intratumoral expression of CHI3L1 correlated with poor survival in 161 lung cancer, 155 pancreatic cancer and 180 colon cancer patients. Conclusions: These results provide the first evidence that Rab37 mediates CHI3L1 secretion in immune cells and highlight nCHI3L1 Abs that can simultaneously target both cancer cells and tumor microenvironment.


Asunto(s)
Proteína 1 Similar a Quitinasa-3/inmunología , Inmunoterapia/métodos , Neoplasias , Proteínas de Unión al GTP rab/inmunología , Animales , Línea Celular Tumoral , Estudios de Cohortes , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Noqueados , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA