Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70.518
Filtrar
1.
Sci Immunol ; 9(95): eadi7418, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758807

RESUMEN

Immune checkpoint blockade is a promising approach to activate antitumor immunity and improve the survival of patients with cancer. V-domain immunoglobulin suppressor of T cell activation (VISTA) is an immune checkpoint target; however, the downstream signaling mechanisms are elusive. Here, we identify leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) as a VISTA binding partner, which acts as an inhibitory receptor by engaging VISTA and suppressing T cell receptor signaling pathways. Mice with T cell-specific LRIG1 deletion developed superior antitumor responses because of expansion of tumor-specific cytotoxic T lymphocytes (CTLs) with increased effector function and survival. Sustained tumor control was associated with a reduction of quiescent CTLs (TCF1+ CD62Lhi PD-1low) and a reciprocal increase in progenitor and memory-like CTLs (TCF1+ PD-1+). In patients with melanoma, elevated LRIG1 expression on tumor-infiltrating CD8+ CTLs correlated with resistance to immunotherapies. These results delineate the role of LRIG1 as an inhibitory immune checkpoint receptor and propose a rationale for targeting the VISTA/LRIG1 axis for cancer immunotherapy.


Asunto(s)
Antígenos B7 , Linfocitos T CD8-positivos , Glicoproteínas de Membrana , Ratones Endogámicos C57BL , Animales , Ratones , Linfocitos T CD8-positivos/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/genética , Humanos , Antígenos B7/inmunología , Antígenos B7/genética , Ratones Noqueados , Línea Celular Tumoral , Femenino , Proteínas de la Membrana , Proteínas del Tejido Nervioso
2.
Sci Immunol ; 9(95): eadi5374, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758808

RESUMEN

The gut microbiota and tumor-associated macrophages (TAMs) affect tumor responses to anti-programmed cell death protein 1 (PD-1) immune checkpoint blockade. Reprogramming TAM by either blocking or deleting the macrophage receptor triggering receptor on myeloid cells 2 (TREM2) attenuates tumor growth, and lack of functional TREM2 enhances tumor elimination by anti-PD-1. Here, we found that anti-PD-1 treatment combined with TREM2 deficiency in mice induces proinflammatory programs in intestinal macrophages and a concomitant expansion of Ruminococcus gnavus in the gut microbiota. Gavage of wild-type mice with R. gnavus enhanced anti-PD-1-mediated tumor elimination, recapitulating the effect occurring in the absence of TREM2. A proinflammatory intestinal environment coincided with expansion, increased circulation, and migration of TNF-producing CD4+ T cells to the tumor bed. Thus, TREM2 remotely controls anti-PD-1 immune checkpoint blockade through modulation of the intestinal immune environment and microbiota, with R. gnavus emerging as a potential probiotic agent for increasing responsiveness to anti-PD-1.


Asunto(s)
Microbioma Gastrointestinal , Inmunoterapia , Macrófagos , Glicoproteínas de Membrana , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1 , Receptores Inmunológicos , Animales , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Ratones , Microbioma Gastrointestinal/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Inmunoterapia/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Macrófagos/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones Noqueados , Femenino , Intestinos/inmunología
3.
ACS Chem Neurosci ; 15(10): 2018-2027, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38701380

RESUMEN

In silico modeling was applied to study the efficiency of two ligands, namely, UCB-J and UCB-F, to bind to isoforms of the synaptic vesicle glycoprotein 2 (SV2) that are involved in the regulation of synaptic function in the nerve terminals, with the ultimate goal to understand the selectivity of the interaction between UCB-J and UCB-F to different isoforms of SV2. Docking and large-scale molecular dynamics simulations were carried out to unravel various binding patterns, types of interactions, and binding free energies, covering hydrogen bonding and nonspecific hydrophobic interactions, water bridge, π-π, and cation-π interactions. The overall preference for bonding types of UCB-J and UCB-F with particular residues in the protein pockets can be disclosed in detail. A unique interaction fingerprint, namely, hydrogen bonding with additional cation-π interaction with the pyridine moiety of UCB-J, could be established as an explanation for its high selectivity over the SV2 isoform A (SV2A). Other molecular details, primarily referring to the presence of π-π interactions and hydrogen bonding, could also be analyzed as sources of selectivity of the UCB-F tracer for the three isoforms. The simulations provide atomic details to support future development of new selective tracers targeting synaptic vesicle glycoproteins and their associated diseases.


Asunto(s)
Glicoproteínas de Membrana , Simulación de Dinámica Molecular , Proteínas del Tejido Nervioso , Isoformas de Proteínas , Ligandos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/química , Humanos , Enlace de Hidrógeno , Unión Proteica/fisiología , Simulación del Acoplamiento Molecular/métodos , Vesículas Sinápticas/metabolismo
4.
J Exp Med ; 221(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38753245

RESUMEN

Preterm infants are at high risk of developing neonatal sepsis. γδ T cells are thought to be an important set of effector cells in neonates. Here, γδ T cells were investigated in a longitudinal cohort of preterm neonates using next-generation sequencing, flow cytometry, and functional assays. During the first year of life, the Vγ9Vδ2 T cell subset showed dynamic phenotypic changes and elevated levels of fetal-derived Vγ9Vδ2 T cells were evident in infants with sepsis. Single-cell transcriptomics identified HLA-DRhiCD83+ γδ T cells in neonatal sepsis, which expressed genes related to antigen presentation. In vitro assays showed that CD83 was expressed on activated Vγ9Vδ2 T cells in preterm and term neonates, but not in adults. In contrast, activation of adult Vγ9Vδ2 T cells enhanced CD86 expression, which was presumably the key receptor to induce CD4 T cell proliferation. Together, we provide a map of the maturation of γδ T cells after preterm birth and highlight their phenotypic diversity in infections.


Asunto(s)
Antígenos CD , Antígeno CD83 , Recien Nacido Prematuro , Receptores de Antígenos de Linfocitos T gamma-delta , Humanos , Recién Nacido , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Recien Nacido Prematuro/inmunología , Antígenos CD/metabolismo , Antígenos CD/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Femenino , Masculino , Sepsis/inmunología , Estudios de Cohortes , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto , Activación de Linfocitos/inmunología , Sepsis Neonatal/inmunología , Lactante
5.
J Cell Mol Med ; 28(10): e18280, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38758159

RESUMEN

Acute lung injury (ALI) is featured with a robust inflammatory response. Angiopoietin-like protein 2 (ANGPTL2), a pro-inflammatory protein, is complicated with various disorders. However, the role of ANGPTL2 in ALI remains to be further explored. The mice and MH-S cells were administrated with lipopolysaccharide (LPS) to evoke the lung injury in vivo and in vitro. The role and mechanism of ANGPTL was investigated by haematoxylin-eosin, measurement of wet/dry ratio, cell count, terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling, reverse transcription quantitative polymerase chain reaction, immunofluorescence, enzyme-linked immunosorbent assay, detection of autophagic flux and western blot assays. The level of ANGPTL2 was upregulated in lung injury. Knockout of ANGPTL2 alleviated LPS-induced pathological symptoms, reduced pulmonary wet/dry weight ratio, the numbers of total cells and neutrophils in BALF, apoptosis rate and the release of pro-inflammatory mediators, and modulated polarization of alveolar macrophages in mice. Knockdown of ANGPTL2 downregulated the level of pyroptosis indicators, and elevated the level of autophagy in LPS-induced MH-S cells. Besides, downregulation of ANGPTL2 reversed the LPS-induced the expression of leukocyte immunoglobulin (Ig)-like receptor B2 (LILRB2) and triggering receptor expressed on myeloid cells 2 (TREM2), which was reversed by the overexpression of LILRB2. Importantly, knockdown of TREM2 reversed the levels of autophagy- and pyroptosis-involved proteins, and the contents of pro-inflammatory factors in LPS-induced MH-S cells transfected with si ANGPTL2, which was further inverted with the treatment of rapamycin. Therefore, ANGPTL2 silencing enhanced autophagy to alleviate alveolar macrophage pyroptosis via reducing LILRB2-mediated inhibition of TREM2.


Asunto(s)
Lesión Pulmonar Aguda , Proteína 2 Similar a la Angiopoyetina , Autofagia , Lipopolisacáridos , Macrófagos Alveolares , Glicoproteínas de Membrana , Piroptosis , Receptores Inmunológicos , Animales , Piroptosis/genética , Piroptosis/efectos de los fármacos , Autofagia/genética , Ratones , Macrófagos Alveolares/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/inducido químicamente , Técnicas de Silenciamiento del Gen , Masculino , Ratones Endogámicos C57BL , Proteínas Similares a la Angiopoyetina/metabolismo , Proteínas Similares a la Angiopoyetina/genética , Ratones Noqueados
6.
Cell Rep Med ; 5(5): 101561, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38744274

RESUMEN

Natural history and mechanisms for persistent cognitive symptoms ("brain fog") following acute and often mild COVID-19 are unknown. In a large prospective cohort of people who underwent testing a median of 9 months after acute COVID-19 in the New York City/New Jersey area, we found that cognitive dysfunction is common; is not influenced by mood, fatigue, or sleepiness; and is correlated with MRI changes in very few people. In a subgroup that underwent cerebrospinal fluid analysis, there are no changes related to Alzheimer's disease or neurodegeneration. Single-cell gene expression analysis in the cerebrospinal fluid shows findings consistent with monocyte recruitment, chemokine signaling, cellular stress, and suppressed interferon response-especially in myeloid cells. Longitudinal analysis shows slow recovery accompanied by key alterations in inflammatory genes and increased protein levels of CXCL8, CCL3L1, and sTREM2. These findings suggest that the prognosis for brain fog following COVID-19 correlates with myeloid-related chemokine and interferon-responsive genes.


Asunto(s)
COVID-19 , Disfunción Cognitiva , SARS-CoV-2 , Análisis de la Célula Individual , Humanos , COVID-19/líquido cefalorraquídeo , COVID-19/patología , COVID-19/complicaciones , Masculino , Análisis de la Célula Individual/métodos , Femenino , Disfunción Cognitiva/líquido cefalorraquídeo , Disfunción Cognitiva/patología , Disfunción Cognitiva/virología , Disfunción Cognitiva/genética , Persona de Mediana Edad , SARS-CoV-2/aislamiento & purificación , Anciano , Receptores Inmunológicos/genética , Estudios Prospectivos , Adulto , Imagen por Resonancia Magnética , Glicoproteínas de Membrana , Interleucina-8
7.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731936

RESUMEN

Multiple myeloma is a malignancy characterized by the accumulation of malignant plasma cells in bone marrow and the production of monoclonal immunoglobulin. A hallmark of cancer is the evasion of immune surveillance. Histone deacetylase inhibitors have been shown to promote the expression of silenced molecules and hold potential to increase the anti-MM efficacy of immunotherapy. The aim of the present work was to assess the potential effect of tinostamustine (EDO-S101), a first-in-class alkylating deacetylase inhibitor, in combination with daratumumab, an anti-CD38 monoclonal antibody (mAb), through different preclinical studies. Tinostamustine increases CD38 expression in myeloma cell lines, an effect that occurs in parallel with an increment in CD38 histone H3 acetylation levels. Also, the expression of MICA and MICB, ligands for the NK cell activating receptor NKG2D, augments after tinostamustine treatment in myeloma cell lines and primary myeloma cells. Pretreatment of myeloma cell lines with tinostamustine increased the sensitivity of these cells to daratumumab through its different cytotoxic mechanisms, and the combination of these two drugs showed a higher anti-myeloma effect than individual treatments in ex vivo cultures of myeloma patients' samples. In vivo data confirmed that tinostamustine pretreatment followed by daratumumab administration significantly delayed tumor growth and improved the survival of mice compared to individual treatments. In summary, our results suggest that tinostamustine could be a potential candidate to improve the efficacy of anti-CD38 mAbs.


Asunto(s)
ADP-Ribosil Ciclasa 1 , Anticuerpos Monoclonales , Mieloma Múltiple , Subfamilia K de Receptores Similares a Lectina de Células NK , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Humanos , ADP-Ribosil Ciclasa 1/metabolismo , ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Ratones , Línea Celular Tumoral , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Glicoproteínas de Membrana/metabolismo , Sinergismo Farmacológico , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Regulación hacia Arriba/efectos de los fármacos
8.
Invest Ophthalmol Vis Sci ; 65(5): 36, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38776115

RESUMEN

Purpose: The purpose of this study was to investigate the protective effect of CD38 deletion on retinal ganglion cells (RGCs) in a mouse retinal ischemia/reperfusion (I/R) model and an optic nerve crush (ONC) model, and to elucidate the underlying molecular mechanisms. Methods: Retinal I/R and ONC models were constructed in mice. PCR was used to identify the deletion of CD38 gene in mice, hematoxylin and eosin (H&E) staining was used to evaluate the changes in retinal morphology, and electroretinogram (ERG) was used to evaluate the changes in retinal function. The survival of RGCs and activation of retinal macroglia were evaluated by immunofluorescence staining. The expression of Sirt1, CD38, Ac-p65, Ac-p53, TNF-α, IL-1ß, and Caspase3 proteins in the retina was further evaluated by protein imprinting. Results: In retinal I/R and ONC models, CD38 deficiency reduced the loss of RGCs and activation of macroglia and protected the retinal function. CD38 deficiency increased the concentration of NAD+, reduced the degree of acetylation of NF-κB p65 and p53, and reduced expression of the downstream inflammatory cytokines TNFα, IL-1ß, and apoptotic protein Caspase3 in the retina in the ONC model. Intraperitoneal injection of the Sirt1 inhibitor EX-527 partially counteracted the effects of CD38 deficiency, suggesting that CD38 deficiency acts at least in part through the NAD+/Sirt1 pathway. Conclusions: CD38 plays an important role in the pathogenesis of retinal I/R and ONC injury. CD38 deletion protects RGCs by attenuating inflammatory responses and apoptosis through the NAD+/Sirt1 pathway.


Asunto(s)
ADP-Ribosil Ciclasa 1 , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , NAD , Traumatismos del Nervio Óptico , Daño por Reperfusión , Células Ganglionares de la Retina , Sirtuina 1 , Animales , Sirtuina 1/metabolismo , Sirtuina 1/genética , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , ADP-Ribosil Ciclasa 1/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Ratones , NAD/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Electrorretinografía , Compresión Nerviosa , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Masculino , Transducción de Señal/fisiología
9.
Genes (Basel) ; 15(5)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38790164

RESUMEN

Cell-free DNA (cfDNA) has recently emerged as a promising minimally invasive diagnostic biomarker for various cancers. In this study, our aim was to identify cfDNA biomarkers by investigating genes that displayed significant differences between glioma patients and their corresponding controls. To accomplish this, we utilized publicly available data from the Gene Expression Omnibus, focusing on 5-hydroxymethylcytosine (5hmC) profiles in both cfDNA and genomic DNA (gDNA) from glioma patients and healthy individuals. The intersection of gene lists derived from these comparative analyses unveiled LRIG1 and ZNF703 as the two genes with elevated 5hmC levels in both the cfDNA of glioma patients and gDNA of glioma tissue compared to their respective controls. The gene expression data revealed both genes were upregulated in glioma tissue compared to normal brain tissue. Integration of 5hmC data revealed a strong positive correlation in the glioma tissue group between 5hmC and the gene expression of the LRIG1 gene. Furthermore, exploration using the AmiCa web tool indicated that LRIG1 gene expression was elevated compared to 17 other cancers included in the database, emphasizing its potential as a distinctive biomarker across multiple cancer types.


Asunto(s)
5-Metilcitosina , Biomarcadores de Tumor , Neoplasias Encefálicas , Ácidos Nucleicos Libres de Células , Glioma , Glicoproteínas de Membrana , Humanos , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Glioma/genética , Glioma/metabolismo , Glioma/patología , Ácidos Nucleicos Libres de Células/genética , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Regulación Neoplásica de la Expresión Génica , Metilación de ADN
10.
J Neuroinflammation ; 21(1): 128, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38745307

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is a progressive neurodegenerative disease of the central nervous system characterized by inflammation-driven synaptic abnormalities. Interleukin-9 (IL-9) is emerging as a pleiotropic cytokine involved in MS pathophysiology. METHODS: Through biochemical, immunohistochemical, and electrophysiological experiments, we investigated the effects of both peripheral and central administration of IL-9 on C57/BL6 female mice with experimental autoimmune encephalomyelitis (EAE), a model of MS. RESULTS: We demonstrated that both systemic and local administration of IL-9 significantly improved clinical disability, reduced neuroinflammation, and mitigated synaptic damage in EAE. The results unveil an unrecognized central effect of IL-9 against microglia- and TNF-mediated neuronal excitotoxicity. Two main mechanisms emerged: first, IL-9 modulated microglial inflammatory activity by enhancing the expression of the triggering receptor expressed on myeloid cells-2 (TREM2) and reducing TNF release. Second, IL-9 suppressed neuronal TNF signaling, thereby blocking its synaptotoxic effects. CONCLUSIONS: The data presented in this work highlight IL-9 as a critical neuroprotective molecule capable of interfering with inflammatory synaptopathy in EAE. These findings open new avenues for treatments targeting the neurodegenerative damage associated with MS, as well as other inflammatory and neurodegenerative disorders of the central nervous system.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Interleucina-9 , Ratones Endogámicos C57BL , Microglía , Sinapsis , Factor de Necrosis Tumoral alfa , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Ratones , Microglía/metabolismo , Microglía/efectos de los fármacos , Microglía/patología , Interleucina-9/metabolismo , Femenino , Factor de Necrosis Tumoral alfa/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Glicoproteínas de Membrana/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Modelos Animales de Enfermedad
11.
Cell Commun Signal ; 22(1): 272, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750472

RESUMEN

BACKGROUND: In the tumor immune microenvironment (TIME), triggering receptor expressed on myeloid cells 2 (trem2) is widely considered to be a crucial molecule on tumor-associated macrophages(TAMs). Multiple studies have shown that trem2 may function as an immune checkpoint in various malignant tumors, mediating tumor immune evasion. However, its specific molecular mechanisms, especially in glioma, remain elusive. METHODS: Lentivirus was transfected to establish cells with stable knockdown of trem2. A Transwell system was used for segregated coculture of glioma cells and microglia. Western blotting, quantitative real-time polymerase chain reaction (qRT‒PCR), and immunofluorescence (IF) were used to measure the expression levels of target proteins. The proliferation, invasion, and migration of cells were detected by colony formation, cell counting kit-8 (CCK8), 5-ethynyl-2'-deoxyuridine (EdU) and transwell assays. The cell cycle, apoptosis rate and reactive oxygen species (ROS) level of cells were assessed using flow cytometry assays. The comet assay and tube formation assay were used to detect DNA damage in glioma cells and angiogenesis activity, respectively. Gl261 cell lines and C57BL/6 mice were used to construct the glioma orthotopic transplantation tumor model. RESULTS: Trem2 was highly overexpressed in glioma TAMs. Knocking down trem2 in microglia suppressed the growth and angiogenesis activity of glioma cells in vivo and in vitro. Mechanistically, knockdown of trem2 in microglia promoted proinflammatory microglia and inhibited anti-inflammatory microglia by activating jak2/stat1 and inhibiting the NF-κB p50 signaling pathway. The proinflammatory microglia produced high concentrations of nitric oxide (NO) and high levels of the proinflammatory cytokines TNF-α, IL-6, and IL-1ß, and caused further DNA damage and promoted the apoptosis rate of tumor cells. CONCLUSIONS: Our findings revealed that trem2 in microglia plays a significant role in the TIME of gliomas. Knockdown of trem2 in microglia might help to improve the efficiency of inhibiting glioma growth and delaying tumor progression and provide new ideas for further treatment of glioma.


Asunto(s)
Glioma , Janus Quinasa 2 , Glicoproteínas de Membrana , Microglía , FN-kappa B , Receptores Inmunológicos , Factor de Transcripción STAT3 , Transducción de Señal , Glioma/genética , Glioma/patología , Glioma/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Microglía/metabolismo , Microglía/patología , Animales , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , FN-kappa B/metabolismo , Ratones , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Línea Celular Tumoral , Ratones Endogámicos C57BL , Técnicas de Silenciamiento del Gen , Proliferación Celular/genética , Humanos , Inflamación/genética , Inflamación/patología , Apoptosis/genética , Progresión de la Enfermedad , Movimiento Celular/genética
12.
J Cell Mol Med ; 28(10): e18368, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38752280

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder of the brain and is manifested by motor and non-motor symptoms because of degenerative changes in dopaminergic neurons of the substantia nigra. PD neuropathology is associated with mitochondrial dysfunction, oxidative damage and apoptosis. Thus, the modulation of mitochondrial dysfunction, oxidative damage and apoptosis by growth factors could be a novel boulevard in the management of PD. Brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin receptor kinase type B (TrkB) are chiefly involved in PD neuropathology. BDNF promotes the survival of dopaminergic neurons in the substantia nigra and enhances the functional activity of striatal neurons. Deficiency of the TrkB receptor triggers degeneration of dopaminergic neurons and accumulation of α-Syn in the substantia nigra. As well, BDNF/TrkB signalling is reduced in the early phase of PD neuropathology. Targeting of BDNF/TrkB signalling by specific activators may attenuate PD neuropathology. Thus, this review aimed to discuss the potential role of BDNF/TrkB activators against PD. In conclusion, BDNF/TrkB signalling is decreased in PD and linked with disease severity and long-term complications. Activation of BDNF/TrkB by specific activators may attenuate PD neuropathology.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Enfermedad de Parkinson , Receptor trkB , Transducción de Señal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Humanos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Receptor trkB/metabolismo , Animales , Glicoproteínas de Membrana/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología
13.
Life Sci ; 348: 122696, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38710279

RESUMEN

The Triggering Receptor Expressed on Myeloid Cells (TREM) family of receptors plays a crucial role in the immune response across various species. Particularly, TREM-1 and TREM-2 have been extensively studied, both in terms of their applications and their expression sites and signaling pathways. However, the same is not observed for the other family members collectively known as TREM-like-transcripts (TREML). The TREML family consists of eight receptors, with TREML1-5 identified in humans and mice, TREML-6 exclusive found in mice, TREML-7 in dogs and horses, and TREML-8 in rabbits and opossums. Despite the limited data available on the TREML members, they have been implicated in different immune and non-immune activities, which have been proposed to display both pro and anti-inflammatory activities, and to influence fundamental biological processes such as coagulation, bone and neurological development. In this review, we have compiled available information regarding the already discovered members of the family and provided foundational framework for understanding the function, localization, and therapeutic potential of all TREML members. Additionally, we hope that this review may shed light on this family of receptors, whose underlying mechanisms are still awaiting elucidation, while emphasizing the need for future studies to explore their functions and potential therapeutic application.


Asunto(s)
Receptores Inmunológicos , Animales , Humanos , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Transducción de Señal , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Receptor Activador Expresado en Células Mieloides 1/genética
14.
J Pak Med Assoc ; 74(5): 852-856, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38783429

RESUMEN

Objective: To determine the expression of podoplanin, and to correlate it with histopathological grades in oral epithelial dysplasia and oral squamous cell carcinoma cases. METHODS: The retrospective, analytical, cross-sectional study was conducted at the City Laboratory, Peshawar, Pakistan, and comprised specimen block data of histologically diagnosed cases of oral benign lesions, dysplastic lesions and oral squamous cell carcinoma from January 2017 to August 2021. Two sections (4um) were cut from each specimen block for Haematoxylin and Eosin staining and immunohistochemistry. The slides were re-evaluated by two pathologists for confirmation of the diagnosis, and podoplanin marker was applied to cases selected using immunohistochemistry. Data was analysed using SPSS 22. RESULTS: Of the 80 cases identified, 68(85%) were analysed. There were 20(29.4%) benign cases; 11(55%) females and 9(45%) males with mean age 39.90±16.23 years, 20(29.4%) oral dysplastic cases; 14(70%) males and 6(30%) females with mean age 57.75±12.02 years, and 28(41.2%) oral squamous cell carcinoma cases; 17(61%) males and 11(39%) females with mean age 50.55±14.80 years. Podoplanin expression in oral epithelial dysplasia cases was significant (p=0.028), while it was not significant in the other 2 groups (p>0.05). CONCLUSIONS: Podoplanin when used along with histopathological evaluation could aid as an adjuvant technique in the diagnosis and grading of oral epithelial dysplasia.


Asunto(s)
Glicoproteínas de Membrana , Neoplasias de la Boca , Humanos , Femenino , Masculino , Glicoproteínas de Membrana/metabolismo , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Persona de Mediana Edad , Adulto , Estudios Transversales , Estudios Retrospectivos , Anciano , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Lesiones Precancerosas/patología , Lesiones Precancerosas/metabolismo , Pakistán/epidemiología , Adulto Joven , Mucosa Bucal/patología , Mucosa Bucal/metabolismo , Clasificación del Tumor , Biomarcadores de Tumor/metabolismo , Inmunohistoquímica
15.
J Agric Food Chem ; 72(20): 11606-11616, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38722802

RESUMEN

In this work, three MP extracts obtained from Torulaspora delbrueckii were added to red wine, and the changes in phenolic composition, color, and astringency were evaluated by HPLC-DAD-ESI-MS, tristimulus colorimetry, and sensory analysis, respectively. The MP extracts modified wine phenolic composition differently depending on the type of MP. Moreover, two MP extracts were able to reduce wine astringency. The fact that the MP-treated wines showed an increased flavanol content suggests the formation of MP-flavanol aggregates that remain in solution. Furthermore, the formation of these aggregates may hinder the interaction of flavanols with salivary proteins in the mouth. The effect of these MPs might be associated with their larger size, which could influence their ability to bind flavanols and salivary proteins. However, one of the astringent-modulating MPs also produced a loss of color, highlighting the importance of assessing the overall impact of MPs on the organoleptic properties of wine.


Asunto(s)
Gusto , Torulaspora , Vino , Vino/análisis , Humanos , Torulaspora/metabolismo , Torulaspora/química , Fenoles/metabolismo , Fenoles/química , Color , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/química , Cromatografía Líquida de Alta Presión , Femenino , Masculino , Glicoproteínas de Membrana
16.
Sci Rep ; 14(1): 11886, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789534

RESUMEN

The E3 ubiquitin-ligase UHRF1 is an epigenetic regulator coordinating DNA methylation and histone modifications. However, little is known about how it regulates adipogenesis or metabolism. In this study, we discovered that UHRF1 is a key regulatory factor for adipogenesis, and we identified the altered molecular pathways that UHRF1 targets. Using CRISPR/Cas9-based knockout strategies, we discovered the whole transcriptomic changes upon UHRF1 deletion. Bioinformatics analyses revealed that key adipogenesis regulators such PPAR-γ and C/EBP-α were suppressed, whereas TGF-ß signaling and fibrosis markers were upregulated in UHRF1-depleted differentiating adipocytes. Furthermore, UHRF1-depleted cells showed upregulated expression and secretion of TGF-ß1, as well as the glycoprotein GPNMB. Treating differentiating preadipocytes with recombinant GPNMB led to an increase in TGF-ß protein and secretion levels, which was accompanied by an increase in secretion of fibrosis markers such as MMP13 and a reduction in adipogenic conversion potential. Conversely, UHRF1 overexpression studies in human cells demonstrated downregulated levels of GPNMB and TGF-ß, and enhanced adipogenic potential. In conclusion, our data show that UHRF1 positively regulates 3T3-L1 adipogenesis and limits fibrosis by suppressing GPNMB and TGF-ß signaling cascade, highlighting the potential relevance of UHRF1 and its targets to the clinical management of obesity and linked metabolic disorders.


Asunto(s)
Adipogénesis , Glicoproteínas de Membrana , Transducción de Señal , Ubiquitina-Proteína Ligasas , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Humanos , Animales , Ratones , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Fibrosis , Factor de Crecimiento Transformador beta/metabolismo , Proteínas del Ojo/metabolismo , Proteínas del Ojo/genética , Células 3T3-L1 , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Adipocitos/metabolismo , Diferenciación Celular
17.
Free Radic Biol Med ; 220: 154-165, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38710340

RESUMEN

BACKGROUND: Liver fibrosis typically develops as a result of chronic liver injury, which involves inflammatory and regenerative processes. The triggering receptor expressed on myeloid cells 2 (TREM2), predominantly expressing in hepatic non-parenchymal cells, plays a crucial role in regulating the function of macrophages. However, its mechanism in liver fibrosis remains poorly defined. METHODS: Experimental liver fibrosis models in wild type and TREM2-/- mice, and in vitro studies with AML-12 cells and Raw264.7 cells were conducted. The expression of TREM2 and related molecular mechanism were evaluated by using samples from patients with liver fibrosis. RESULTS: We demonstrated that TREM2 was upregulated in murine model with liver fibrosis. Mice lacking TREM2 exhibited reduced phagocytosis activity in macrophages following carbon tetrachloride (CCl4) intoxication. As a result, there was an increased accumulation of necrotic apoptotic hepatocytes. Additionally, TREM2 knockout aggravated the release of mitochondrial damage-associated molecular patterns (mito-DAMPs) from dead hepatocytes during CCl4 exposure, and further promoted the occurrence of macrophage-mediated M1 polarization. Then, TREM2-/- mice showed more serious fibrosis pathological changes. In vitro, the necrotic apoptosis inhibitor GSK872 effectively alleviated the release of mito-DAMPs in AML-12 cells after CCl4 intoxication, which confirmed that mito-DAMPs originated from dead liver cells. Moreover, direct stimulation of Raw264.7 cells by mito-DAMPs from liver tissue can induce intracellular inflammatory response. More importantly, TREM2 was elevated and inflammatory factors were markedly accumulated surrounding dead cells in the livers of human patients with liver fibrosis. CONCLUSION: Our study highlights that TREM2 serves as a negative regulator of liver fibrosis, suggesting its potential as a novel therapeutic target.


Asunto(s)
Hepatocitos , Inflamación , Cirrosis Hepática , Macrófagos , Glicoproteínas de Membrana , Ratones Noqueados , Receptores Inmunológicos , Animales , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Ratones , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Humanos , Hepatocitos/metabolismo , Hepatocitos/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/genética , Células RAW 264.7 , Macrófagos/metabolismo , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Tetracloruro de Carbono/toxicidad , Masculino , Ratones Endogámicos C57BL , Apoptosis , Fagocitosis , Mitocondrias/metabolismo , Mitocondrias/patología , Modelos Animales de Enfermedad
18.
Cell Mol Life Sci ; 81(1): 231, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38780647

RESUMEN

CD200 is an anti-inflammatory protein that facilitates signal transduction through its receptor, CD200R, in cells, resulting in immune response suppression. This includes reducing M1-like macrophages, enhancing M2-like macrophages, inhibiting NK cell cytotoxicity, and downregulating CTL responses. Activation of CD200R has been found to modulate dendritic cells, leading to the induction or enhancement of Treg cells expressing Foxp3. However, the precise mechanisms behind this process are still unclear. Our previous study demonstrated that B cells in Peyer's patches can induce Treg cells, so-called Treg-of-B (P) cells, through STAT6 phosphorylation. This study aimed to investigate the role of CD200 in Treg-of-B (P) cell generation. To clarify the mechanisms, we used wild-type, STAT6 deficient, and IL-24 deficient T cells to generate Treg-of-B (P) cells, and antagonist antibodies (anti-CD200 and anti-IL-20RB), an agonist anti-CD200R antibody, CD39 inhibitors (ARL67156 and POM-1), a STAT6 inhibitor (AS1517499), and soluble IL-20RB were also applied. Our findings revealed that Peyer's patch B cells expressed CD200 to activate the CD200R on T cells and initiate the process of Treg-of-B (P) cells generation. CD200 and CD200R interaction triggers the phosphorylation of STAT6, which regulated the expression of CD200R, CD39, and IL-24 in T cells. CD39 regulated the expression of IL-24, which sustained the expression of CD223 and IL-10 and maintained the cell viability. In summary, the generation of Treg-of-B (P) cells by Peyer's patch B cells was through the CD200R-STAT6-CD39-IL-24 axis pathway.


Asunto(s)
Linfocitos B , Factor de Transcripción STAT6 , Linfocitos T Reguladores , Animales , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Ratones , Linfocitos B/inmunología , Linfocitos B/metabolismo , Factor de Transcripción STAT6/metabolismo , Ratones Endogámicos C57BL , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Antígenos CD/metabolismo , Antígenos CD/genética , Antígenos CD/inmunología , Transducción de Señal , Fosforilación , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/citología , Apirasa/metabolismo , Apirasa/inmunología , Glicoproteínas de Membrana
19.
CNS Neurosci Ther ; 30(5): e14778, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38801174

RESUMEN

AIMS: Synaptic vesicle protein 2A (SV2A) is a unique therapeutic target for pharmacoresistant epilepsy (PRE). As seizure-induced neuronal programmed death, parthanatos was rarely reported in PRE. Apoptosis-inducing factor (AIF), which has been implicated in parthanatos, shares a common cytoprotective function with SV2A. We aimed to investigate whether parthanatos participates in PRE and is mitigated by SV2A via AIF. METHODS: An intraperitoneal injection of lithium chloride-pilocarpine was used to establish an epileptic rat model, and phenytoin and phenobarbital sodium were utilized to select PRE and pharmacosensitive rats. The expression of SV2A was manipulated via lentivirus delivery into the hippocampus. Video surveillance was used to assess epileptic ethology. Biochemical tests were employed to test hippocampal tissues following a successful SV2A infection. Molecular dynamic calculations were used to simulate the interaction between SV2A and AIF. RESULTS: Parthanatos core index, PARP1, PAR, nuclear AIF and MIF, γ-H2AX, and TUNEL staining were all increased in PRE. SV2A is bound to AIF to form a stable complex, successfully inhibiting AIF and MIF nuclear translocation and parthanatos and consequently mitigating spontaneous recurrent seizures in PRE. Moreover, parthanatos deteriorated after the SV2A reduction. SIGNIFICANCE: SV2A protected hippocampal neurons and mitigated epileptic seizures by inhibiting parthanatos via binding to AIF in PRE.


Asunto(s)
Factor Inductor de la Apoptosis , Modelos Animales de Enfermedad , Epilepsia Refractaria , Glicoproteínas de Membrana , Proteínas del Tejido Nervioso , Ratas Sprague-Dawley , Animales , Ratas , Factor Inductor de la Apoptosis/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Epilepsia Refractaria/metabolismo , Epilepsia Refractaria/tratamiento farmacológico , Glicoproteínas de Membrana/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Anticonvulsivantes/farmacología
20.
Cell Biol Toxicol ; 40(1): 40, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38797732

RESUMEN

MYBL1 is a strong transcriptional activator involved in the cell signaling. However, there is no systematic study on the role of MYBL1 in atherosclerosis. The aim of this study is to elucidate the role and mechanism of MYBL1 in atherosclerosis. GSE28829, GSE43292 and GSE41571 were downloaded from NCBI for differentially expressed analysis. The expression levels of MYBL1 in atherosclerotic plaque tissue and normal vessels were detected by qRT-PCR, Western blot and Immunohistochemistry. Transwell and CCK-8 were used to detect the migration and proliferation of HUVECs after silencing MYBL1. RNA-seq, Western blot, qRT-PCR, Luciferase reporter system, Immunofluorescence, Flow cytometry, ChIP and CO-IP were used to study the role and mechanism of MYBL1 in atherosclerosis. The microarray data of GSE28829, GSE43292, and GSE41571 were analyzed and intersected, and then MYBL1 were verified. MYBL1 was down-regulated in atherosclerotic plaque tissue. After silencing of MYBL1, HUVECs were damaged, and their migration and proliferation abilities were weakened. Overexpression of MYBL1 significantly enhanced the migration and proliferation of HUVECs. MYBL1 knockdown induced abnormal autophagy in HUVEC cells, suggesting that MYBL1 was involved in the regulation of HUVECs through autophagy. Mechanistic studies showed that MYBL1 knockdown inhibited autophagosome and lysosomal fusion in HUVECs by inhibiting PLEKHM1, thereby exacerbating atherosclerosis. Furthermore, MYBL1 was found to repress lipid accumulation in HUVECs after oxLDL treatment. MYBL1 knockdown in HUVECs was involved in atherosclerosis by inhibiting PLEKHM1-induced autophagy, which provided a novel target of therapy for atherosclerosis.


Asunto(s)
Aterosclerosis , Autofagia , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Aterosclerosis/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Autofagia/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proliferación Celular/genética , Movimiento Celular/genética , Regulación hacia Abajo/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Transactivadores/metabolismo , Transactivadores/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA