Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26.744
Filtrar
1.
Food Res Int ; 186: 114338, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729719

RESUMEN

Women with the extremely prevalent polycystic ovary syndromegather multiple cardiovascular risk factors and chronic subclinical inflammation. Interactions between diet, adiposity, and gut microbiota modulate intestinal permeabilityand bacterial product translocation, and may contribute to the chronic inflammation process associated with the polycystic ovary syndrome. In the present study, we aimed to address the effects of obesity, functional hyperandrogenism, and diverse oral macronutrients on intestinal permeabilityby measuring circulating markers of gut barrier dysfunction and endotoxemia. Participants included 17 non-hyperandrogenic control women, 17 women with polycystic ovary syndrome, and 19 men that were submitted to glucose, lipid, and protein oral loads. Lipopolysaccharide-binding protein, plasma soluble CD14, succinate, zonulin family peptide, and glucagon-like peptide-2 were determined at fasting and after oral challenges. Macronutrient challenges induced diverse changes on circulating intestinal permeabilitybiomarkers in the acute postprancial period, with lipids and proteins showing the most unfavorable and favorable effects, respectively. Particularly, lipopolysaccharide-binding protein, zonulin family peptide, and glucagon-like peptide-2 responses were deregulated by the presence of obesity after glucose and lipid challenges. Obese subjects showed higher fasting intestinal permeabilitybiomarkers levels than non-obese individuals, except for plasma soluble CD14. The polycystic ovary syndromeexacerbated the effect of obesity further increasing fasting glucagon-like peptide-2, lipopolysaccharide-binding protein, and succinate concentrations. We observed specific interactions of the polycystic ovary syndromewith obesity in the postprandial response of succinate, zonulin family peptide, and glucagon-like peptide-2. In summary, obesity and polycystic ovary syndromemodify the effect of diverse macronutrients on the gut barrier, and alsoinfluence intestinal permeabilityat fasting,contributing to the morbidity of functional hyperandrogenism by inducing endotoxemia and subclinical chronic inflammation.


Asunto(s)
Ayuno , Péptido 2 Similar al Glucagón , Obesidad , Permeabilidad , Síndrome del Ovario Poliquístico , Humanos , Síndrome del Ovario Poliquístico/metabolismo , Femenino , Adulto , Ayuno/sangre , Masculino , Péptido 2 Similar al Glucagón/sangre , Mucosa Intestinal/metabolismo , Microbioma Gastrointestinal , Nutrientes , Adulto Joven , Haptoglobinas/metabolismo , Endotoxemia , Receptores de Lipopolisacáridos/sangre , Proteínas de Fase Aguda/metabolismo , Biomarcadores/sangre , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/metabolismo , Grasas de la Dieta , Glucosa/metabolismo , Funcion de la Barrera Intestinal , Proteínas Portadoras , Precursores de Proteínas
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Mol Neurodegener ; 19(1): 41, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38760857

RESUMEN

Recent evidence suggests that Alzheimer's disease (AD) genetic risk variants (rs1582763 and rs6591561) of the MS4A locus are genome-wide significant regulators of soluble TREM2 levels such that the minor allele of the protective variant (rs1582763) is associated with higher sTREM2 and lower AD risk while the minor allele of (rs6591561) relates to lower sTREM2 and higher AD risk. Our group previously found that higher sTREM2 relates to higher Aß40, worse blood-brain barrier (BBB) integrity (measured with the CSF/plasma albumin ratio), and higher CSF tau, suggesting strong associations with amyloid abundance and both BBB and neurodegeneration complicate interpretation. We expand on this work by leveraging these common variants as genetic tools to tune the interpretation of high CSF sTREM2, and by exploring the potential modifying role of these variants on the well-established associations between CSF sTREM2 as well as TREM2 transcript levels in the brain with AD neuropathology. Biomarker analyses leveraged data from the Vanderbilt Memory & Aging Project (n = 127, age = 72 ± 6.43) and were replicated in the Alzheimer's Disease Neuroimaging Initiative (n = 399, age = 73 ± 7.39). Autopsy analyses were performed leveraging data from the Religious Orders Study and Rush Memory and Aging Project (n = 577, age = 89 ± 6.46). We found that the protective variant rs1582763 attenuated the association between CSF sTREM2 and Aß40 (ß = -0.44, p-value = 0.017) and replicated this interaction in ADNI (ß = -0.27, p = 0.017). We did not observe this same interaction effect between TREM2 mRNA levels and Aß peptides in brain (Aß total ß = -0.14, p = 0.629; Aß1-38, ß = 0.11, p = 0.200). In contrast to the effects on Aß, the minor allele of this same variant seemed to enhance the association with blood-brain barrier dysfunction (ß = 7.0e-4, p = 0.009), suggesting that elevated sTREM2 may carry a much different interpretation in carriers vs. non-carriers of this allele. When evaluating the risk variant (rs6591561) across datasets, we did not observe a statistically significant interaction against any outcome in VMAP and observed opposing directions of associations in ADNI and ROS/MAP on Aß levels. Together, our results suggest that the protective effect of rs1582763 may act by decoupling the associations between sTREM2 and amyloid abundance, providing important mechanistic insight into sTREM2 changes and highlighting the need to incorporate genetic context into the analysis of sTREM2 levels, particularly if leveraged as a clinical biomarker of disease in the future.


Asunto(s)
Enfermedad de Alzheimer , Biomarcadores , Glicoproteínas de Membrana , Receptores Inmunológicos , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Anciano , Masculino , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/metabolismo , Femenino , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/líquido cefalorraquídeo , Anciano de 80 o más Años , Encéfalo/metabolismo , Encéfalo/patología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Predisposición Genética a la Enfermedad
9.
Sci Adv ; 10(18): eadn6537, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38701219

RESUMEN

In mammals, males and females show marked differences in immune responses. Males are globally more sensitive to infectious diseases, while females are more susceptible to systemic autoimmunity. X-chromosome inactivation (XCI), the epigenetic mechanism ensuring the silencing of one X in females, may participate in these sex biases. We perturbed the expression of the trigger of XCI, the noncoding RNA Xist, in female mice. This resulted in reactivation of genes on the inactive X, including members of the Toll-like receptor 7 (TLR7) signaling pathway, in monocyte/macrophages and dendritic and B cells. Consequently, female mice spontaneously developed inflammatory signs typical of lupus, including anti-nucleic acid autoantibodies, increased frequencies of age-associated and germinal center B cells, and expansion of monocyte/macrophages and dendritic cells. Mechanistically, TLR7 signaling is dysregulated in macrophages, leading to sustained expression of target genes upon stimulation. These findings provide a direct link between maintenance of XCI and female-biased autoimmune manifestations and highlight altered XCI as a cause of autoimmunity.


Asunto(s)
Autoinmunidad , Macrófagos , Receptor Toll-Like 7 , Inactivación del Cromosoma X , Animales , Femenino , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Autoinmunidad/genética , Ratones , Masculino , Macrófagos/metabolismo , Macrófagos/inmunología , ARN Largo no Codificante/genética , Transducción de Señal , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología
10.
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
11.
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
12.
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
13.
Commun Biol ; 7(1): 541, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714838

RESUMEN

Age-related diseases pose great challenges to health care systems worldwide. During aging, endothelial senescence increases the risk for cardiovascular disease. Recently, it was described that Phosphatase 1 Nuclear Targeting Subunit (PNUTS) has a central role in cardiomyocyte aging and homeostasis. Here, we determine the role of PNUTS in endothelial cell aging. We confirm that PNUTS is repressed in senescent endothelial cells (ECs). Moreover, PNUTS silencing elicits several of the hallmarks of endothelial aging: senescence, reduced angiogenesis and loss of barrier function. Findings are validate in vivo using endothelial-specific inducible PNUTS-deficient mice (Cdh5-CreERT2;PNUTSfl/fl), termed PNUTSEC-KO. Two weeks after PNUTS deletion, PNUTSEC-KO mice present severe multiorgan failure and vascular leakage. Transcriptomic analysis of PNUTS-silenced HUVECs and lungs of PNUTSEC-KO mice reveal that the PNUTS-PP1 axis tightly regulates the expression of semaphorin 3B (SEMA3B). Indeed, silencing of SEMA3B completely restores barrier function after PNUTS loss-of-function. These results reveal a pivotal role for PNUTS in endothelial homeostasis through a SEMA3B downstream pathway that provides a potential target against the effects of aging in ECs.


Asunto(s)
Senescencia Celular , Células Endoteliales de la Vena Umbilical Humana , Ratones Noqueados , Semaforinas , Animales , Ratones , Humanos , Semaforinas/metabolismo , Semaforinas/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales/metabolismo , Envejecimiento/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Ratones Endogámicos C57BL , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Endotelio Vascular/metabolismo
14.
Sci Transl Med ; 16(741): eadj9052, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38569016

RESUMEN

Microglia help limit the progression of Alzheimer's disease (AD) by constraining amyloid-ß (Aß) pathology, effected through a balance of activating and inhibitory intracellular signals delivered by distinct cell surface receptors. Human leukocyte Ig-like receptor B4 (LILRB4) is an inhibitory receptor of the immunoglobulin (Ig) superfamily that is expressed on myeloid cells and recognizes apolipoprotein E (ApoE) among other ligands. Here, we find that LILRB4 is highly expressed in the microglia of patients with AD. Using mice that accumulate Aß and carry a transgene encompassing a portion of the LILR region that includes LILRB4, we corroborated abundant LILRB4 expression in microglia wrapping around Aß plaques. Systemic treatment of these mice with an anti-human LILRB4 monoclonal antibody (mAb) reduced Aß load, mitigated some Aß-related behavioral abnormalities, enhanced microglia activity, and attenuated expression of interferon-induced genes. In vitro binding experiments established that human LILRB4 binds both human and mouse ApoE and that anti-human LILRB4 mAb blocks such interaction. In silico modeling, biochemical, and mutagenesis analyses identified a loop between the two extracellular Ig domains of LILRB4 required for interaction with mouse ApoE and further indicated that anti-LILRB4 mAb may block LILRB4-mApoE by directly binding this loop. Thus, targeting LILRB4 may be a potential therapeutic avenue for AD.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Humanos , Ratones , Animales , Microglía/metabolismo , Anticuerpos/metabolismo , Receptores de Superficie Celular/metabolismo , Amiloide/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E , Leucocitos/metabolismo , Ratones Transgénicos , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo
15.
JCI Insight ; 9(7)2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38587072

RESUMEN

The roles of fibronectin leucine-rich transmembrane protein 2 (FLRT2) in physiological and pathological processes are not well known. Here, we identify a potentially novel function of FLRT2 in preventing endothelial cell senescence and vascular aging. We found that FLRT2 expression was lower in cultured senescent endothelial cells as well as in aged rat and human vascular tissues. FLRT2 mediated endothelial cell senescence via the mTOR complex 2, AKT, and p53 signaling pathway in human endothelial cells. We uncovered that FLRT2 directly associated with integrin subunit beta 4 (ITGB4) and thereby promoted ITGB4 phosphorylation, while inhibition of ITGB4 substantially mitigated the induction of senescence triggered by FLRT2 depletion. Importantly, FLRT2 silencing in mice promoted vascular aging, and overexpression of FLRT2 rescued a premature vascular aging phenotype. Therefore, we propose that FLRT2 could be targeted therapeutically to prevent senescence-associated vascular aging.


Asunto(s)
Células Endoteliales , Proteína p53 Supresora de Tumor , Animales , Humanos , Ratones , Ratas , Envejecimiento , Células Endoteliales/metabolismo , Integrina beta4/genética , Integrina beta4/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Glicoproteínas de Membrana/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
16.
Nat Commun ; 15(1): 3213, 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38615060

RESUMEN

Oxidative stress-induced lipid accumulation is mediated by lipid droplets (LDs) homeostasis, which sequester vulnerable unsaturated triglycerides into LDs to prevent further peroxidation. Here we identify the upregulation of lipopolysaccharide-binding protein (LBP) and its trafficking through LDs as a mechanism for modulating LD homeostasis in response to oxidative stress. Our results suggest that LBP induces lipid accumulation by controlling lipid-redox homeostasis through its lipid-capture activity, sorting unsaturated triglycerides into LDs. N-acetyl-L-cysteine treatment reduces LBP-mediated triglycerides accumulation by phospholipid/triglycerides competition and Peroxiredoxin 4, a redox state sensor of LBP that regulates the shuttle of LBP from LDs. Furthermore, chronic stress upregulates LBP expression, leading to insulin resistance and obesity. Our findings contribute to the understanding of the role of LBP in regulating LD homeostasis and against cellular peroxidative injury. These insights could inform the development of redox-based therapies for alleviating oxidative stress-induced metabolic dysfunction.


Asunto(s)
Proteínas de Fase Aguda , Gotas Lipídicas , Glicoproteínas de Membrana , Proteínas de Fase Aguda/metabolismo , Proteínas Portadoras/metabolismo , Homeostasis , Gotas Lipídicas/metabolismo , Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/metabolismo , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Triglicéridos
17.
EBioMedicine ; 103: 105098, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38608514

RESUMEN

BACKGROUND: The widespread involvement of tumor-infiltrating B cells highlights their potential role in tumor behavior. However, B cell heterogeneity in PDAC remains unexplored. Studying TIL-Bs in PDAC aims to identify new treatment strategies. METHODS: We performed single-cell RNA sequencing to study the heterogeneity of B cells in PDAC. The prognostic and immunologic value of the identified CD38+ B cells was explored in FUSCC (n = 147) and TCGA (n = 176) cohorts. Flow cytometry was conducted to characterize the relationship between CD38+ B cells and other immune cells, as well as their phenotypic features. In vitro and in vivo experiments were performed to assess the putative effect of CD38+ B cells on antitumor immunity. FINDINGS: The presence of CD38+ B cells in PDAC was associated with unfavorable clinicopathological features and poorer overall survival (p < 0.001). Increased infiltration of CD38+ B cells was accompanied by reduced natural killer (NK) cells (p = 0.021) and increased regulatory T cells (p = 0.016). Molecular profiling revealed high expression of IL-10, IL-35, TGF-ß, GZMB, TIM-1, CD5 and CD21, confirming their putative regulatory B cell-like features. Co-culture experiments demonstrated suppression of NK cell cytotoxicity by CD38+ B cell-derived IL-10 (p < 0.001). Finally, in vivo experiments suggested adoptive transfer of CD38+ B cells reduced antitumor immunity and administration of a CD38 inhibitor hampered tumor growth (p < 0.001). INTERPRETATION: We discovered regulatory B cell-like CD38+ B cell infiltration as an independent prognostic factor in PDAC. The use of CD38 inhibitor may provide new possibilities for PDAC immunotherapy. FUNDING: This study was supported by the National Natural Science Foundation of China (U21A20374), Shanghai Municipal Science and Technology Major Project (21JC1401500), Scientific Innovation Project of Shanghai Education Committee (2019-01-07-00-07-E00057), Special Project for Clinical Research in the Health Industry of the Shanghai Health Commission (No. 20204Y0265) and Natural Science Foundation of Shanghai (23ZR1479300).


Asunto(s)
ADP-Ribosil Ciclasa 1 , Carcinoma Ductal Pancreático , Humanos , ADP-Ribosil Ciclasa 1/metabolismo , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/terapia , Animales , Ratones , Pronóstico , Antígenos CD19/metabolismo , Antígenos CD19/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Femenino , Masculino , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Línea Celular Tumoral , Microambiente Tumoral/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Persona de Mediana Edad , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Terapia de Inmunosupresión
18.
Mol Neurodegener ; 19(1): 37, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38654375

RESUMEN

BACKGROUND: Microglia play important roles in maintaining brain homeostasis and neurodegeneration. The discovery of genetic variants in genes predominately or exclusively expressed in myeloid cells, such as Apolipoprotein E (APOE) and triggering receptor expressed on myeloid cells 2 (TREM2), as the strongest risk factors for Alzheimer's disease (AD) highlights the importance of microglial biology in the brain. The sequence, structure and function of several microglial proteins are poorly conserved across species, which has hampered the development of strategies aiming to modulate the expression of specific microglial genes. One way to target APOE and TREM2 is to modulate their expression using antisense oligonucleotides (ASOs). METHODS: In this study, we identified, produced, and tested novel, selective and potent ASOs for human APOE and TREM2. We used a combination of in vitro iPSC-microglia models, as well as microglial xenotransplanted mice to provide proof of activity in human microglial in vivo. RESULTS: We proved their efficacy in human iPSC microglia in vitro, as well as their pharmacological activity in vivo in a xenografted microglia model. We demonstrate ASOs targeting human microglia can modify their transcriptional profile and their response to amyloid-ß plaques in vivo in a model of AD. CONCLUSIONS: This study is the first proof-of-concept that human microglial can be modulated using ASOs in a dose-dependent manner to manipulate microglia phenotypes and response to neurodegeneration in vivo.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Oligonucleótidos Antisentido , Microglía/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Humanos , Oligonucleótidos Antisentido/farmacología , Animales , Ratones , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Células Madre Pluripotentes Inducidas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Modelos Animales de Enfermedad
19.
Metabolism ; 155: 155911, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38609037

RESUMEN

BACKGROUND: The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing year by year and has become one of the leading causes of end-stage liver disease worldwide. Triggering Receptor Expressed on Myeloid Cells 2 (Trem2) has been confirmed to play an essential role in the progression of MASLD, but its specific mechanism still needs to be clarified. This study aims to explore the role and mechanism of Trem2 in MASLD. METHODS: Human liver tissues were obtained from patients with MASLD and controls. Myeloid-specific knockout mice (Trem2mKO) and myeloid-specific overexpression mice (Trem2TdT) were fed a high-fat diet, either AMLN or CDAHFD, to establish the MASLD model. Relevant signaling molecules were assessed through lipidomics and RNA-seq analyses after that. RESULTS: Trem2 is upregulated in human MASLD/MASH-associated macrophages and is associated with hepatic steatosis and inflammation progression. Hepatic steatosis and inflammatory responses are exacerbated with the knockout of myeloid Trem2 in MASLD mice, while mice overexpressing Trem2 exhibit the opposite phenomenon. Mechanistically, Trem2mKO can aggravate macrophage pyroptosis through the PI3K/AKT signaling pathway and amplify the resulting inflammatory response. At the same time, Trem2 promotes the inflammation resolution phenotype transformation of macrophages through TGFß1, thereby promoting tissue repair. CONCLUSIONS: Myeloid Trem2 ameliorates the progression of Metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. We believe targeting myeloid Trem2 could represent a potential avenue for treating MASLD.


Asunto(s)
Progresión de la Enfermedad , Hígado Graso , Inflamación , Macrófagos , Glicoproteínas de Membrana , Ratones Noqueados , Piroptosis , Receptores Inmunológicos , Animales , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Humanos , Macrófagos/metabolismo , Inflamación/metabolismo , Inflamación/patología , Piroptosis/fisiología , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/genética , Masculino , Ratones Endogámicos C57BL , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Enfermedades Metabólicas/genética , Hígado/metabolismo , Hígado/patología
20.
J Immunol ; 212(11): 1680-1692, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38607278

RESUMEN

Plasmacytoid dendritic cells (pDCs) are strongly implicated as a major source of IFN-I in systemic lupus erythematosus (SLE), triggered through TLR-mediated recognition of nucleic acids released from dying cells. However, relatively little is known about how TLR signaling and IFN-I production are regulated in pDCs. In this article, we describe a role for integrin αvß3 in regulating TLR responses and IFN-I production by pDCs in mouse models. We show that αv and ß3-knockout pDCs produce more IFN-I and inflammatory cytokines than controls when stimulated through TLR7 and TLR9 in vitro and in vivo. Increased cytokine production was associated with delayed acidification of endosomes containing TLR ligands, reduced LC3 conjugation, and increased TLR signaling. This dysregulated TLR signaling results in activation of B cells and promotes germinal center (GC) B cell and plasma cell expansion. Furthermore, in a mouse model of TLR7-driven lupus-like disease, deletion of αvß3 from pDCs causes accelerated autoantibody production and pathology. We therefore identify a pDC-intrinsic role for αvß3 in regulating TLR signaling and preventing activation of autoreactive B cells. Because αvß3 serves as a receptor for apoptotic cells and cell debris, we hypothesize that this regulatory mechanism provides important contextual cues to pDCs and functions to limit responses to self-derived nucleic acids.


Asunto(s)
Autoinmunidad , Células Dendríticas , Integrina alfaVbeta3 , Lupus Eritematoso Sistémico , Ratones Noqueados , Transducción de Señal , Receptor Toll-Like 7 , Animales , Ratones , Células Dendríticas/inmunología , Integrina alfaVbeta3/inmunología , Integrina alfaVbeta3/metabolismo , Autoinmunidad/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 7/genética , Lupus Eritematoso Sistémico/inmunología , Transducción de Señal/inmunología , Ratones Endogámicos C57BL , Citocinas/metabolismo , Citocinas/inmunología , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo , Linfocitos B/inmunología , Autoanticuerpos/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Activación de Linfocitos/inmunología , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA