Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
FEBS Lett ; 596(21): 2834-2850, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36053046

RESUMEN

The hypoxia-inducible factors (HIF)-1α and HIF-2α are central regulators of transcriptional programmes in settings such as development and tumour expansion. HIF-2α moonlights as a cap-dependent translation factor. We provide new insights into how the interferon-stimulated gene 15 (ISG15), a ubiquitin-like modifier, and the HIFs regulate one another in hypoxia and interferon-induced cells. We show that upon ISGylation induction and HIF-α stabilization, both HIFs promote protein ISGylates through transcriptional and/or post-transcriptional pathways. We show the first evidence of HIF-2α modification by ISG15. ISGylation induces system-level alterations to the HIF transcriptional programme and increases the cytoplasmic/nuclear fraction and translation activity of HIF-2α. This work identifies ISG15 as a regulator of hypoxic mRNA translation, which has implications for immune processes and disease progression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Hipoxia , Polirribosomas , Humanos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Hipoxia de la Célula/genética , Hipoxia de la Célula/inmunología , Hipoxia/genética , Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Interferones/genética , Interferones/inmunología , Polirribosomas/genética , Polirribosomas/inmunología
2.
Cancer Lett ; 528: 76-84, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34973392

RESUMEN

Cancer cells acquire chemoresistance in hypoxic regions of solid tumors, which is suggested to be at least partly due to reduction of their proliferative activity. However, molecular mechanisms behind it have not been fully elucidated. Here, we revealed the importance of active proteolysis of a histone acetylation reader, ATPase family AAA domain containing 2 (ATAD2), under hypoxia. We found that inactivation of an O2/Fe2+/α-ketoglutarate-dependent dioxygenase triggered ATAD2 proteolysis by the proteasome system upon severe hypoxia in a hypoxia-inducible factors (HIFs)-independent manner. Consistently, ATAD2 expression levels were markedly lower in perinecrotic hypoxic regions in both xenografted and clinical tumor tissues. The ATAD2 proteolysis was accompanied by a decrease in the amount of acetylated histone H3 lysine 27 and inhibited cell cycle progression from the early to late S phase under severe hypoxia. The retardation of S phase progression induced chemoresistance, which was blocked by overexpression of ATAD2. Together, these results indicate that ATAD2 proteolysis upon severe hypoxia induces chemoresistance of cancer cells through heterochromatinization and the subsequent retardation of S phase progression; therefore, inhibition of ATAD2 proteolysis is expected to be a strategy to overcome chemoresistance of hypoxic tumor cells.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Ciclo Celular/inmunología , Hipoxia de la Célula/inmunología , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Acetilación , Humanos , Proteolisis , Fase S , Transfección
3.
Cell Death Dis ; 12(11): 1068, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753919

RESUMEN

Systemic transplantation of oxygen-glucose deprivation (OGD)-preconditioned primary microglia enhances neurological recovery in rodent stroke models, albeit the underlying mechanisms have not been sufficiently addressed. Herein, we analyzed whether or not extracellular vesicles (EVs) derived from such microglia are the biological mediators of these observations and which signaling pathways are involved in the process. Exposing bEnd.3 endothelial cells (ECs) and primary cortical neurons to OGD, the impact of EVs from OGD-preconditioned microglia on angiogenesis and neuronal apoptosis by the tube formation assay and TUNEL staining was assessed. Under these conditions, EV treatment stimulated both angiogenesis and tube formation in ECs and repressed neuronal cell injury. Characterizing microglia EVs by means of Western blot analysis and other techniques revealed these EVs to be rich in TGF-ß1. The latter turned out to be a key compound for the therapeutic potential of microglia EVs, affecting the Smad2/3 pathway in both ECs and neurons. EV infusion in stroke mice confirmed the aforementioned in vitro results, demonstrating an activation of the TGF-ß/Smad2/3 signaling pathway within the ischemic brain. Furthermore, enriched TGF-ß1 in EVs secreted from OGD-preconditioned microglia stimulated M2 polarization of residing microglia within the ischemic cerebral environment, which may contribute to a regulation of an early inflammatory response in postischemic hemispheres. These observations are not only interesting from the mechanistic point of view but have an immediate therapeutic implication as well, since stroke mice treated with such EVs displayed a better functional recovery in the behavioral test analyses. Hence, the present findings suggest a new way of action of EVs derived from OGD-preconditioned microglia by regulating the TGF-ß/Smad2/3 pathway in order to promote tissue regeneration and neurological recovery in stroke mice.


Asunto(s)
Hipoxia de la Célula/inmunología , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Microglía/metabolismo , Neovascularización Patológica/metabolismo , Accidente Cerebrovascular/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Apoptosis , Humanos , Ratones , Accidente Cerebrovascular/patología , Transfección
4.
Front Immunol ; 12: 727508, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603299

RESUMEN

Several in vitro cellular models have been developed with the aim to reproduce and dissect human granulomatous responses, the hallmark of tuberculosis (TB) immunopathogenesis. In that context, we compared two- (2D) versus three-dimensional (3D) granuloma models resulting from infection of human peripheral blood mononuclear cells with M. tuberculosis (Mtb) in the absence or presence of a collagen-based extracellular matrix (ECM). Granuloma formation was found to be significantly enhanced in the 2D model. This feature was associated with an earlier chemokine production and lymphocyte activation, but also a significantly increased bacterial burden. Remarkably, the reduction in Mtb burden in the 3D model correlated with an increase in GM-CSF production. GM-CSF, which is known to promote macrophage survival, was found to be inherently induced by the ECM. We observed that only 3D in vitro granulomas led to the accumulation of lipid inclusions within Mtb. Our data suggest that a hypoxic environment within the ECM could be responsible for this dormant-like Mtb phenotype. Furthermore, exposure to a TNF-α antagonist reverted Mtb dormancy, thereby mimicking the reactivation of TB observed in rheumatic patients receiving this therapy. To conclude, we showed that only in vitro granulomas generated in the presence of an ECM could recapitulate some clinically relevant features of granulomatous responses in TB. As such, this model constitutes a highly valuable tool to study the interplay between immunity and Mtb stress responses as well as to evaluate novel treatment strategies.


Asunto(s)
Hipoxia de la Célula/inmunología , Matriz Extracelular/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Granuloma/inmunología , Mycobacterium tuberculosis , Agregación Celular , Células Cultivadas , Humanos , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Fagocitosis , Especies Reactivas de Oxígeno/inmunología
5.
Eur J Pharmacol ; 910: 174500, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34509462

RESUMEN

Angiogenesis is an early and key event in the pathogenesis of rheumatoid arthritis (RA) and is crucial for the proliferation of synovial tissue and the formation of pannus. This process is regulated by both angiogenesis-stimulating factors and angiogenesis inhibitors, the basis for the "on-off hypothesis of angiogenesis." In RA, inflammation, immune imbalance, and hypoxia can further turn on the switch for blood vessel formation and induce angiogenesis. The new vasculature can recruit white blood cells, induce immune imbalance, and aggravate inflammation. At the same time, it also can provide oxygen and nutrients for the proliferating synovial tissue, which can accelerate the process of RA. The current therapies for RA mainly target the inflammatory response of autoimmune activation. Although these therapies have been greatly improved, there are still many patients whose RA is difficult to treat or who do not fully respond to treatment. Therefore, new innovative therapies are still urgently needed. This review covers the mechanism of synovial angiogenesis in RA, including the detailed process of angiogenesis and the relationship between inflammation, immune imbalance, hypoxia, and synovial angiogenesis, respectively. At the same time, in the context of the development of angiogenesis inhibition therapy for cancer, we also discuss similar treatment strategies for RA, especially the combination of targeted angiogenesis inhibition therapy and immunotherapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Artritis Reumatoide/tratamiento farmacológico , Factores Inmunológicos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Membrana Sinovial/efectos de los fármacos , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Hipoxia de la Célula/inmunología , Quimioterapia Combinada/métodos , Humanos , Factores Inmunológicos/uso terapéutico , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Membrana Sinovial/irrigación sanguínea , Membrana Sinovial/inmunología , Membrana Sinovial/patología
6.
Int Immunopharmacol ; 98: 107915, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34198236

RESUMEN

Non-alcoholic steatohepatitis (NASH), an extreme progressive subtype of metabolic associated fatty liver disease, is well characterized by hepatic steatosis, injury and inflammation. It causes irreversible hepatic damage and there are no approved interventions for it. ß-PAE, a representatively pharmacological active substance isolated from Pogostemon cablin, has been indicated to alleviate hepatic steatosis and injury through modulating lipid metabolism in rats with simple steatosis. However, its protection against NASH remains unclear. Here, this study explored the potential effect of ß-PAE against high-fat diet-induced NASH in rats. The results displayed that ß-PAE significantly reduced the gains of body weight and epididymal adipose tissue, liver index and attenuated liver histological damages in NASH rats. It also markedly alleviated hepatic inflammation by inhibiting NLRP3 inflammasome activation. In NASH, the active NLRP3 inflammasome is caused by hepatic lipid abnormal accumulation-induced oxidative stress. Excessive oxidative stress results in hepatic histanoxia, which exacerbates lipid metabolism disorders by elevating CD36 to suppress AMPK signalling pathways. Moreover, the lipid accumulation led by lipid metabolism dysfunction intensifies oxidative stress. A vicious circle is formed among oxidative stress, histanoxia and lipid accumulation, eventually, but ß-PAE effectively interrupted it. Interestingly, soluble CD36 (sCD36) was tightly associated not only with hepatic steatosis and injury but also with inflammation. Collectively, ß-PAE exerted a positive effect against NASH by interrupting the vicious circle among oxidative stress, histanoxia and lipid accumulation, and sCD36 may be a promising non-invasive tool for NASH diagnosis.


Asunto(s)
Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Sesquiterpenos de Guayano/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/inmunología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/inmunología , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Ratas , Sesquiterpenos de Guayano/uso terapéutico
7.
J Immunol Res ; 2021: 1815098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34307691

RESUMEN

Adiponectin is a small peptide secreted and a key component of the endocrine system and immune system. Although globular adiponectin protects myocardial ischemia/reperfusion-induced cardiomyocyte injury, the protective mechanisms remain largely unresolved. Using a neonatal rat ventricular myocyte hypoxia/reoxygenation model, we investigated the role of its potential mechanisms of necroptosis in globular adiponectin-mediated protection in hypoxia/reoxygenation-induced cardiomyocyte injury as compared to apoptosis. We found that globular adiponectin treatment attenuated cardiomyocyte injury as indicated by increased cell viability and reduced lactate dehydrogenase release following hypoxia/reoxygenation. Immunofluorescence staining and Western blotting demonstrated that both necroptosis and apoptosis were triggered by hypoxia/reoxygenation and diminished by globular adiponectin. Necrostatin-1 (RIP1-specific inhibitor) and Z-VAD-FMK (pan-caspase inhibitor) only mimicked the inhibition of necroptosis and apoptosis, respectively, by globular adiponectin in hypoxia/reoxygenation-treated cardiomyocytes. Globular adiponectin attenuated reactive oxygen species production, oxidative damage, and p38MAPK and NF-κB signaling, all important for necroptosis and apoptosis. Collectively, our study suggests that globular adiponectin inhibits hypoxia/reoxygenation-induced necroptosis and apoptosis in cardiomyocytes probably by reducing oxidative stress and interrupting p38MAPK signaling.


Asunto(s)
Adiponectina/metabolismo , Daño por Reperfusión Miocárdica/inmunología , Miocitos Cardíacos/patología , Animales , Animales Recién Nacidos , Apoptosis/inmunología , Hipoxia de la Célula/inmunología , Supervivencia Celular , Células Cultivadas , Medios de Cultivo/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/inmunología , Necroptosis/inmunología , Estrés Oxidativo/inmunología , Embarazo , Cultivo Primario de Células , Ratas , Especies Reactivas de Oxígeno/metabolismo
8.
J Immunol Res ; 2021: 9979843, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34307696

RESUMEN

Ischemic heart disease is a leading cause of mortality and morbidity worldwide. We previously demonstrated that acacetin protects against myocardial ischemia reperfusion injury in rats, although the underlying mechanism remains to be elucidated. In the present study, we investigated the effects of acacetin on autophagy during hypoxia/reoxygenation (H/R) injury by exposing H9c2 myocardial cells to H/R with or without acacetin pretreatment during hypoxia. Our results show that acacetin significantly increased cell viability in a dose-dependent manner, enhanced antioxidant capacity, and suppressed protein apoptosis of rat cardiomyocytes H9c2 cells following H/R injury. In addition, lentiviral infection of H9c2 cardiomyocytes revealed that acacetin pretreatment significantly enhanced the fluorescence intensity of autophagy proteins Beclin 1, LC3-II, and p62. These results indicate that acacetin protected H9c2 cardiomyocytes from H/R damage by enhancing autophagy. Moreover, we found that application of acacetin increased activation of the PI3K/Akt signaling pathway, whereas cotreatment with the PI3K inhibitor LY294002 reversed the inhibition of apoptosis and autophagy induced by acacetin. In conclusion, acacetin mitigated H/R injury by promoting autophagy through activating the PI3K/Akt/mTOR signaling pathway.


Asunto(s)
Flavonas/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Autofagia/efectos de los fármacos , Autofagia/inmunología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/inmunología , Línea Celular , Cromonas/farmacología , Modelos Animales de Enfermedad , Flavonas/uso terapéutico , Humanos , Morfolinas/farmacología , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
9.
Mol Pharm ; 18(8): 2935-2946, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34191525

RESUMEN

The adoptive transfer of natural killer (NK) cells, which can recognize and obliterate cancer cells, provides a practical alternative to current treatment modalities to improve cancer patients' survival. However, translating NK cell therapies to treat solid tumors has proven challenging due to the tumor microenvironment (TME). Hypoxia in the TME induces immunosuppression that inhibits the cytotoxic function of NK cells. Thus, reversing hypoxia-induced immunosuppression is critical for effective adoptive NK cell immunotherapy. In this study, we use manganese dioxide nanoparticles (MnO2 NPs) to catalyze the degradation of tumor-produced hydrogen peroxide, thereby generating oxygen. For improved biocompatibility and modulation of oxygen production, the MnO2 NPs were encapsulated into poly(lactic-co-glycolic) to produce particles that are 116 nm in size and with a ζ-potential of +17 mV (PLGA-MnO2 NPs). The PLGA-MnO2 NPs showed first-order oxygen production and sustained high oxygen tension compared to equivalent amounts of bare MnO2 NPs in the presence of H2O2. The PLGA-MnO2 NPs were biocompatible, reduced hypoxia after penetration into the core of cancer spheroids, and decreased hypoxia-induced factor 1 α expression. Reducing hypoxia in the spheroid resulted in a decrease in the potent immunosuppressors, adenosine, and lactate, which was confirmed by electrospray ionization mass spectroscopy (ESI-MS). ESI-MS also showed a change in the metabolism of the amino acids aspartate, glutamine, and glutamate after hypoxia reduction in the cancer cells. Notably, the spheroids' microenvironment changes enhanced NK cells' cytotoxicity, which obliterated the spheroids. These results demonstrate that reducing hypoxia-induced immunosuppression in tumors is a potent strategy to increase the potency of cytotoxic immune cells in the TME. The developed NPs are promising new tools to improve adoptive NK cell therapy.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Células Asesinas Naturales/inmunología , Compuestos de Manganeso/farmacología , Nanopartículas del Metal/química , Nanocápsulas/química , Óxidos/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Esferoides Celulares/inmunología , Adenosina/metabolismo , Traslado Adoptivo/métodos , Neoplasias de la Mama/patología , Hipoxia de la Célula/inmunología , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Ácido Láctico/metabolismo , Células MCF-7 , Compuestos de Manganeso/química , Óxidos/química , Tamaño de la Partícula , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
10.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34108245

RESUMEN

Patients with severe COVID-19 infection exhibit a low level of oxygen in affected tissue and blood. To understand the pathophysiology of COVID-19 infection, it is therefore necessary to understand cell function during hypoxia. We investigated aspects of human monocyte activation under hypoxic conditions. HMGB1 is an alarmin released by stressed cells. Under normoxic conditions, HMGB1 activates interferon regulatory factor (IRF)5 and nuclear factor-κB in monocytes, leading to expression of type I interferon (IFN) and inflammatory cytokines including tumor necrosis factor α, and interleukin 1ß, respectively. When hypoxic monocytes are activated by HMGB1, they produce proinflammatory cytokines but fail to produce type I IFN. Hypoxia-inducible factor-1α, induced by hypoxia, functions as a direct transcriptional repressor of IRF5 and IRF3. As hypoxia is a stressor that induces secretion of HMGB1 by epithelial cells, hypoxia establishes a microenvironment that favors monocyte production of inflammatory cytokines but not IFN. These findings have implications for the pathogenesis of COVID-19.


Asunto(s)
Hipoxia de la Célula/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Monocitos/inmunología , COVID-19/inmunología , Células Cultivadas , Citocinas/inmunología , Humanos , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interleucina-1beta/metabolismo , Monocitos/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Oxígeno/metabolismo , SARS-CoV-2/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Cancer Res ; 81(15): 4001-4013, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33990397

RESUMEN

In pancreatic cancer, the robust fibroinflammatory stroma contributes to immune suppression and renders tumors hypoxic, altering intratumoral metabolic pathways and leading to poor survival. One metabolic enzyme activated during hypoxia is lactate dehydrogenase A (LDHA). As a result of its promiscuous activity under hypoxia, LDHA produces L-2 hydroxyglutarate (L-2HG), an epigenetic modifier, that regulates the tumor transcriptome. However, the role of L-2HG in remodeling the pancreatic tumor microenvironment is not known. Here we used mass spectrometry to detect L-2HG in serum samples from patients with pancreatic cancer, comprising tumor cells as well as stromal cells. Both hypoxic pancreatic tumors as well as serum from patients with pancreatic cancer accumulated L-2HG as a result of promiscuous activity of LDHA. This abnormally accumulated L-2HG led to H3 hypermethylation and altered gene expression, which regulated a critical balance between stemness and differentiation in pancreatic tumors. Secreted L-2HG inhibited T-cell proliferation and migration, suppressing antitumor immunity. In a syngeneic orthotopic model of pancreatic cancer, inhibition of LDH with GSK2837808A decreased L-2HG, induced tumor regression, and sensitized tumors to anti-PD1 therapy. In conclusion, hypoxia-mediated promiscuous activity of LDH produces L-2HG in pancreatic tumor cells, regulating the stemness-differentiation balance and contributing to immune evasion. Targeting LDH can be developed as a potential therapy to sensitize pancreatic tumors to checkpoint inhibitor therapy. SIGNIFICANCE: This study shows that promiscuous LDH activity produces L-2HG in pancreatic tumor and stromal cells, modulating tumor stemness and immune cell function and infiltration in the tumor microenvironment.


Asunto(s)
Hipoxia de la Célula/inmunología , Evasión Inmune/inmunología , Neoplasias Pancreáticas/inmunología , Animales , Diferenciación Celular , Femenino , Humanos , Ratones , Transfección
12.
Front Immunol ; 12: 641883, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33927716

RESUMEN

Cellular metabolism of both cancer and immune cells in the acidic, hypoxic, and nutrient-depleted tumor microenvironment (TME) has attracted increasing attention in recent years. Accumulating evidence has shown that cancer cells in TME could outcompete immune cells for nutrients and at the same time, producing inhibitory products that suppress immune effector cell functions. Recent progress revealed that metabolites in the TME could dysregulate gene expression patterns in the differentiation, proliferation, and activation of immune effector cells by interfering with the epigenetic programs and signal transduction networks. Nevertheless, encouraging studies indicated that metabolic plasticity and heterogeneity between cancer and immune effector cells could provide us the opportunity to discover and target the metabolic vulnerabilities of cancer cells while potentiating the anti-tumor functions of immune effector cells. In this review, we will discuss the metabolic impacts on the immune effector cells in TME and explore the therapeutic opportunities for metabolically enhanced immunotherapy.


Asunto(s)
Diferenciación Celular/inmunología , Proliferación Celular , Epigénesis Genética/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias , Microambiente Tumoral/inmunología , Animales , Hipoxia de la Célula/inmunología , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neoplasias/metabolismo
13.
Int Immunopharmacol ; 95: 107509, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33761438

RESUMEN

B cells play a major role in regulating disease incidence through various factors, including spleen tyrosine kinase (Syk), which transmits signals to all hematopoietic lineage cells. Hypoxia-inducible factor (HIF)-1α accumulates under hypoxic conditions, which is also oxidative stress to induce nuclear factor (erythroid-derived 2)-like 2 (Nrf2) responsible for gene expression of antioxidant enzymes. In the present study, we investigated whether B cells are regulated by crosstalk of HIF-1α and Nrf2 via reactive oxygen species (ROS)-mediated Syk activation. When B cells were incubated under hypoxic conditions, Syk phosphorylation, HIF-1α, and Nrf2 levels were increased. Hypoxia-inducible results were consistent with CoCl2 treatment, which mimics hypoxic conditions. Cell viability was reduced by the Syk inhibitor BAY 61-3606. Increased Nrf2 levels due to hypoxia or CoCl2 were inhibited by treatment with a HIF inhibitor. Hypoxia- or CoCl2-induced ROS increased HIF-1α and Nrf2 levels, which were attenuated by treatment with N-acetyl-L-cysteine (NAC), a ROS scavenger. HIF-1α levels were reduced in doxycycline-treated shNrf2 cells. Clobetasol propionate, a Nrf2 inhibitor, also inhibited HIF-1α levels induced by hypoxia or CoCl2. ROS-mediated Syk phosphorylation at tyrosine 525/526 was confirmed by treatment with H2O2, hypoxia, and CoCl2, and attenuated with NAC treatment. Inhibition of Syk phosphorylation by BAY 61-3606 is consistent with a decrease in protein HIF-1α and Nrf2 levels. Taken together, HIF-1α levels might control Nrf2 levels and vice versa, and could be associated with Syk phosphorylation in B cells. The results indicate that B cells could be regulated by crosstalk of HIF-1α and Nrf2 through ROS-mediated Syk activation.


Asunto(s)
Linfocitos B/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Factor 2 Relacionado con NF-E2/inmunología , Quinasa Syk/inmunología , Animales , Hipoxia de la Célula/inmunología , Línea Celular , Supervivencia Celular , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/inmunología
14.
Front Immunol ; 12: 630430, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679775

RESUMEN

C-reactive protein (CRP) is the best-known acute phase protein. In humans, almost every type of inflammation is accompanied by an increase of CRP concentration. Until recently, the only known physiological function of CRP was the marking of cells to initiate their phagocytosis. This triggers the classical complement pathway up to C4, which helps to eliminate pathogens and dead cells. However, vital cells with reduced energy supply are also marked, which is useful in the case of a classical external wound because an important substrate for pathogens is disposed of, but is counterproductive at internal wounds (e.g., heart attack or stroke). This mechanism negatively affects clinical outcomes since it is established that CRP levels correlate with the prognosis of these indications. Here, we summarize what we can learn from a clinical study in which CRP was adsorbed from the bloodstream by CRP-apheresis. Recently, it was shown that CRP can have a direct effect on blood pressure in rabbits. This is interesting in regard to patients with high inflammation, as they often become tachycardic and need catecholamines. These two physiological effects of CRP apparently also occur in COVID-19. Parts of the lung become ischemic due to intra-alveolar edema and hemorrhage and in parallel CRP increases dramatically, hence it is assumed that CRP is also involved in this ischemic condition. It is meanwhile considered that most of the damage in COVID-19 is caused by the immune system. The high amounts of CRP could have an additional influence on blood pressure in severe COVID-19.


Asunto(s)
Proteína C-Reactiva/inmunología , COVID-19/inmunología , Infarto del Miocardio/inmunología , SARS-CoV-2/inmunología , Accidente Cerebrovascular/inmunología , Animales , Muerte Celular/inmunología , Hipoxia de la Célula/inmunología , Complemento C4/inmunología , Humanos , Conejos
15.
Int Immunopharmacol ; 94: 107456, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33588175

RESUMEN

OBJECTIVE: To investigate the effects of hypoxia and Porphyromonas gingivalis- lipopolysaccharide (P. gingivalis-LPS) on activation of the NACHT leucine-rich repeat protein 3 (NLRP3) inflammasome in human gingival fibroblasts (HGFs). DESIGN: Periodontitis was optimally simulated using a hypoxic concentration of 1%. HGFs were stimulated using P. gingivalis-LPS (1.0 µg/ml) in normoxia and hypoxia for 3 h and 6 h, respectively. The expression levels of genes and proteins of hypoxia-inducible factor-1α (HIF-1α), interleukin-1ß, gasdermin D (GSDMD) and the NLRP3 inflammasome, including NLRP3, apoptosis-associated speck-like protein containing CARD (ASC), caspase-1 and its activated forms, were measured using quantitative real-time polymerase chain reaction and western blot. ELISA was used to detect and determine levels of the inflammatory factor interleukin-1ß in cell supernatants. Lactate dehydrogenase (LDH) release assay, caspase-1 activity assay and Hoechst 33342/Propidium Iodide (PI) staining were performed to further verify the presence of pyroptosis. RESULTS: The NLRP3 inflammasome (i.e., NLRP3, ASC, caspase-1) was not affected by individual stimulation using P. gingivalis-LPS or hypoxia. However, the combination of both hypoxia and P. gingivalis-LPS stimulation significantly enhanced inflammasome activation and promoted the expression of interleukin-1ß, gasdermin D and HIF-1α at gene and protein levels; PI positive cells and the release of LDH were also elevated. CONCLUSION: Hypoxia and P. gingivalis-LPS synergistically induced NLRP3 inflammasome activation in HGFs, and subsequently high levels of interleukin-1ß and GSDMD-mediated pyroptosis can cause an HGF inflammatory response, which plays an important role in the pathogenesis of periodontitis.


Asunto(s)
Hipoxia de la Célula/inmunología , Fibroblastos/inmunología , Inflamasomas/inmunología , Lipopolisacáridos , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Porphyromonas gingivalis , Adolescente , Adulto , Femenino , Encía/citología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/inmunología , Adulto Joven
16.
J Reprod Immunol ; 144: 103281, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33549904

RESUMEN

MiR-126-3p is a prototype of an endothelial miRNA and has protective effects on endothelial cells. However, little is known about the effects of miR-126-3p on placental trophoblasts. In the present study, we tested the hypothesis that aberrant miR-126-3p expression is present in preeclamptic placenta which contributes to increased inflammatory response in trophoblasts. Placentas were obtained immediately after delivery from normotensive and preeclamptic pregnancies. Villous tissue was either fixed with formalin or used for trophoblast isolation. Trophoblast miR-126-3p expression was assessed by in situ hybridization of formalin-fixed tissue sections and by RT-PCR in cultured syncytiotrophoblasts. Culture medium was collected for measurement of IL-6, TNFα, and 8-Isoprostane production by ELISA and total cellular protein was collected for evaluation of HIF1α expression by Western blot. Effects of overexpression of miR-126-3p in trophoblasts on cytokine production were tested by transfection of pre-mir-126, a precursor of miR-126, into primary isolated trophoblasts. We found that downregulation of miR-126-3p expression was associated with increased IL-6 and TNFα production in trophoblasts from preeclamptic placentas vs. normal placentas. Moreover, transient overexpression of miR-126-3p significantly reduced IL-6 and TNFα production in trophoblasts from both normal and preeclamptic placentas. We further found that increase in miR-126-3p expression not only suppressed hypoxia-induced increases in IL-6 and TNFα production, but also attenuated hypoxia-induced increases in HIF1α expression and 8-Isoprostane production in trophoblasts cultured under hypoxic condition. These results provide plausible evidence that downregulation of miR-126-3p expression reduces anti-inflammatory and anti-oxidative stress activities in placental trophoblasts in preeclampsia.


Asunto(s)
Regulación hacia Abajo/inmunología , MicroARNs/metabolismo , Preeclampsia/inmunología , Trofoblastos/patología , Adulto , Hipoxia de la Célula/inmunología , Células Cultivadas , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-6/genética , MicroARNs/genética , Estrés Oxidativo/genética , Estrés Oxidativo/inmunología , Preeclampsia/patología , Embarazo , Trofoblastos/inmunología , Trofoblastos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Adulto Joven
17.
Cell Death Dis ; 12(1): 95, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33462215

RESUMEN

Intestinal ischemia-reperfusion (IR) injury is associated with high mortality rates, which have not improved in the past decades despite advanced insight in its pathophysiology using in vivo animal and human models. The inability to translate previous findings to effective therapies emphasizes the need for a physiologically relevant in vitro model to thoroughly investigate mechanisms of IR-induced epithelial injury and test potential therapies. In this study, we demonstrate the use of human small intestinal organoids to model IR injury by exposing organoids to hypoxia and reoxygenation (HR). A mass-spectrometry-based proteomics approach was applied to characterize organoid differentiation and decipher protein dynamics and molecular mechanisms of IR injury in crypt-like and villus-like human intestinal organoids. We showed successful separation of organoids exhibiting a crypt-like proliferative phenotype, and organoids exhibiting a villus-like phenotype, enriched for enterocytes and goblet cells. Functional enrichment analysis of significantly changing proteins during HR revealed that processes related to mitochondrial metabolism and organization, other metabolic processes, and the immune response were altered in both organoid phenotypes. Changes in protein metabolism, as well as mitophagy pathway and protection against oxidative stress were more pronounced in crypt-like organoids, whereas cellular stress and cell death associated protein changes were more pronounced in villus-like organoids. Profile analysis highlighted several interesting proteins showing a consistent temporal profile during HR in organoids from different origin, such as NDRG1, SDF4 or DMBT1. This study demonstrates that the HR response in human intestinal organoids recapitulates properties of the in vivo IR response. Our findings provide a framework for further investigations to elucidate underlying mechanisms of IR injury in crypt and/or villus separately, and a model to test therapeutics to prevent IR injury.


Asunto(s)
Hipoxia de la Célula/inmunología , Intestinos/fisiopatología , Organoides/fisiopatología , Proteómica/métodos , Daño por Reperfusión/fisiopatología , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos
18.
J Mater Chem B ; 9(2): 336-348, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33242320

RESUMEN

Integration of various therapeutic modes and novel hypoxic therapy are two emerging aspects in the current anti-cancer field. Based on this, we designed a multifunctional therapeutic system combining photothermal therapy (PTT), the newly defined chemodynamic therapy (CDT) and AIPH-based hypoxic therapy ingeniously, which can take effect well in hypoxic tumor environments. The CuFeSe2-based heterojunction was controllably constructed by the coating of a MIL-100(Fe) shell layer by layer, and the large mesoporous cavities were subsequently filled with a polymerization initiator (AIPH) and phase change material (tetradecanol) to achieve higher drug loading and controlled heat release of radicals. When irradiated by a single 808 nm laser, the photothermal agent of CuFeSe2 plays a significant role of the initiating switch in the whole nanoplatform, whose hyperthermia not only realizes fundamental PTT but also promotes greatly the Fenton reaction of the MIL-100(Fe) shell for oxidative ˙OH production and the generation of toxic AIPH radicals while melting tetradecanol. Due to the sensitive heat-responsive therapies independent of oxygen concentration, the nanoplatform showed a superior therapeutic effect for hypoxic tumor environments. Besides, on account of the effective attenuation for X-rays and the presence of the magnetic element Fe of CuFeSe2, the nanoplatform was also certified to be a superior diagnosis agent for computed tomography (CT) and magnetic resonance imaging (MRI). As expected, cell experiments in vitro and mice experiments in vivo further verified the excellent biocompatibility and antitumor effect, suggesting that this nanoplatform of CuFeSe2@MIL-100(Fe)-AIPH is promising for simultaneous diagnosis and treatment in hypoxic cancer therapy.


Asunto(s)
Hipoxia de la Célula/inmunología , Rayos Infrarrojos/uso terapéutico , Nanoestructuras/química , Neoplasias/terapia , Fotoquimioterapia/métodos , Humanos
19.
Theranostics ; 10(25): 11479-11496, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33052227

RESUMEN

Tubular damage initiated by inflammatory response and ischemic/hypoxic stress is a hallmark of septic acute kidney injury (AKI), albeit the molecular mechanism coupling the two events remains unclear. We investigated the intrinsic nature of tubular damage with respect to inflammatory/hypoxic stress during septic AKI. Methods: The apoptotic response of tubular cells to LPS stimuli was analyzed before and after hypoxia exposure. Cellular ubiquitination, co-immunoprecipitation, GST-pulldown, in vitro protein kinase assay, immunofluorescence and CRISPR technology were adopted to determine the molecular mechanism underlying this process. In vivo characterization was performed in wild-type and DAPK1-/- mice models of cecal ligation and puncture (CLP). Results: We found that the MyD88-dependent inflammatory response couples to tubular damage during LPS stimuli under hypoxia in a Fn14/SCFFbxw7α-dispensable manner via recruitment of caspase-8 with TRIF-RIP1 signalosome mediated by DAPK1, which directly binds to and phosphorylates Pellino1 at Ser39, leading to Pellino1 poly-ubiquitination and turnover. Either pharmacological deactivation or genetic ablation of DAPK1 makes tubular cells refractory to the LPS-induced damage in the context of hypoxia, while kinase activity of DAPK1 is essential for ruin execution. Targeting DAPK1 effectively protects mice against septic AKI and potentiates the efficacy of a MyD88 homodimerization inhibitor, ST2825. Conclusion: Our findings provide a rationale for the mechanism whereby inflammation intersects with hypoxic tubular damage during septic AKI through a previously unappreciated role of DAPK1-inducible Ser39 phosphorylation in Pellino1 turnover and underscore that combined targeting DAPK1 and MyD88 might be a feasible strategy for septic AKI management.


Asunto(s)
Lesión Renal Aguda/inmunología , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Proteínas Nucleares/metabolismo , Sepsis/complicaciones , Ubiquitina-Proteína Ligasas/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Sistemas CRISPR-Cas/genética , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/inmunología , Línea Celular , Proteínas Quinasas Asociadas a Muerte Celular/antagonistas & inhibidores , Proteínas Quinasas Asociadas a Muerte Celular/genética , Modelos Animales de Enfermedad , Células Epiteliales , Técnicas de Inactivación de Genes , Compuestos Heterocíclicos con 2 Anillos/farmacología , Compuestos Heterocíclicos con 2 Anillos/uso terapéutico , Humanos , Túbulos Renales/citología , Túbulos Renales/patología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/antagonistas & inhibidores , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas Nucleares/genética , Oxidación-Reducción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Células RAW 264.7 , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Compuestos de Espiro/farmacología , Compuestos de Espiro/uso terapéutico , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
20.
Biosci Rep ; 40(10)2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-32990315

RESUMEN

BACKGROUND: Tongxinluo (TXL) capsule, a polypharmacy derived from traditional Chinese medicine (TCM), has been widely used in coronary heart disease (CHD), while the underlying mechanism of TXL capsule is still unclear. The present study aimed at investigating the underlying mechanism of TXL acting on CHD patients and providing substantial evidence in molecular evidence by means of a network pharmacological analysis. METHOD: Active compounds and targeted genes of TXL were retrieved from TCM systems pharmacology (TCMSP) and TCM integrative database (TCMID). CHD and coronary artery disease were treated as search queries in GeneCards and Online Mendelian Inheritance in Man (OMIM) databases to obtain disease-related genes. Visualization of disease-targets network was performed under administration of Cytoscape software. Besides, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were administered. H9c2 cells were used to validate the predicted results in cardiomyocytes/reoxygenation model, and anti-inflammatory ability was examined. RESULTS: A network of a total of 212 nodes and 1016 edges was obtained. Peptide and ubiquitin-like protein ligase binding occupied a leading position of GO enrichment. For KEGG analysis, fluid shear stress and atherosclerosis, as well as inflammation-related pathways were enriched. Cellular validation revealed the anti-inflammatory effect of ß-sitosterol, eriodictyol, odoricarpin, and tirucallol as active compounds of TXL. CONCLUSION: Our study provided substantial molecular evidence that TXL capsule possessed the characteristics of multitargets with safe profile, and the main component is capable of regulating cytokine level in CHD patients.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedad Coronaria/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Redes Reguladoras de Genes/efectos de los fármacos , Animales , Antiinflamatorios/uso terapéutico , Hipoxia de la Célula/inmunología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Enfermedad Coronaria/genética , Enfermedad Coronaria/inmunología , Citocinas/análisis , Citocinas/inmunología , Citocinas/metabolismo , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...