Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
MedComm (2020) ; 5(6): e610, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38881675

RESUMEN

Noncanonical pyroptosis is triggered by Caspase 4/5/11, which cleaves Gasdermin D (GSDMD), leading to cell lysis. While GSDMD has been studied previously in systemic lupus erythematosus (SLE), the role of pyroptosis in SLE pathogenesis remains unclear and contentious, with limited understanding of Caspase 11-mediated pyroptosis in this condition. In this study, we explored the level of Caspase 11-mediated pyroptosis in SLE, identifying both the upstream pathways and the interaction between pyroptosis and adaptive immune responses. We observed increased Caspase 5/11 and GSDMD-dependent pyroptosis in the macrophages/monocytes of both lupus patients and mice. We identified serum lipopolysaccharide (LPS), released from the gut due to a compromised gut barrier, as the signal that triggers Caspase 11 activation in MRL/lpr mice. We further discovered that pyroptotic macrophages promote the differentiation of mature B cells independently of T cells. Additionally, inhibiting Caspase 11 and preventing LPS leakage proved effective in improving lupus symptoms in MRL/lpr mice. These findings suggest that elevated serum LPS, resulting from a damaged gut barrier, induces Caspase 11/GSDMD-mediated pyroptosis, which in turn promotes B cell differentiation and enhances autoimmune responses in SLE. Thus, targeting Caspase 11 could be a viable therapeutic strategy for SLE.

2.
Theranostics ; 14(3): 1049-1064, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38250043

RESUMEN

Rationale: Macrophage-associated inflammation and keratinocytes excessive proliferation and inflammatory cytokines secretion induced by stimulation play an important role in the progression of psoriasiform dermatitis. However, how these two types of cells communicate remains obscure. Methods: We induced a mouse model with experimental psoriasiform dermatitis by Imiquimod (IMQ). To investigate whether damaged keratinocytes promote macrophage polarization and accelerate skin lesions by releasing extracellular vesicle (EV), purified EV were isolated from the primary epidermis of 5-day IMQ-induced psoriasiform dermatitis model mice, and then fluorescence-labeled the EV with PKH67. The EV was injected into the skin of mice treated with IMQ or vehicle 2 days in situ. In addition, we established a co-culture system of the human monocytic cell line (THP-1) and HaCaT, and THP-1/HaCaT conditioned media culture model in vitro respectively. Subsequently, we evaluated the effect of Leucine-rich α-2-glycoprotein 1 (LRG1)-enriched EV on macrophage activation. Results: We demonstrated macrophages can significantly promote keratinocyte inflammation and macrophage polarization may be mediated by intercellular communication with keratinocytes. Interestingly, IMQ-induced 5-day, keratinocyte-derived EV recruited macrophage and enhanced the progression of skin lesions. Similar to results in vivo, EV released from M5-treated HaCaT significantly promotes Interleukin 1ß (IL-1ß) and Tumor necrosis factor α (TNF-α) expression of THP-1 cells. Importantly, we found that LRG1-enriched EV regulates macrophages via TGF beta Receptor 1 (TGFßR1) dependent process. Conclusion: Our findings indicated a novel mechanism for promoting psoriasiform dermatitis, which could be a potential therapeutic target.


Asunto(s)
Dermatitis , Vesículas Extracelulares , Humanos , Animales , Ratones , Queratinocitos , Macrófagos , Glicoproteínas , Inflamación
3.
J Autoimmun ; 141: 103047, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37179169

RESUMEN

Systemic lupus erythematosus (SLE) is a highly heterogeneous autoimmune disease characterized by multiple organ damage accompanied by the over-production of autoantibodies. Decreased intestinal flora diversity and disruption of homeostasis have been proven to be associated with pathogenesis of SLE. In previous study, a clinical trial was conducted to verify the safety and effectiveness of fecal microbiota transplantation (FMT) in the treatment of SLE. To explore the mechanism of FMT in the treatment of SLE, we included 14 SLE patients participating in clinical trials, including 8 in responders group (Rs) and 6 in non-responders group (NRs), and collected peripheral blood DNA and serum. We found that the serum of S-adenosylmethionine (SAM), methylation group donor, was upregulated after FMT, accompanied by an increase in genome-wide DNA methylation level in Rs. We further showed that the methylation levels in promoter regions of Interferon-γ (IFN-γ), induced Helicase C Domain Containing Protein 1 (IFIH1), endoplasmic reticulum membrane protein complex 8 (EMC8), and Tripartite motif-containing protein 58 (TRIM58) increased after FMT treatment. On the contrary, there was no significant change in the methylation of IFIH1 promoter region in the NRs after FMT, and the methylation level of IFIH1 in the Rs was significantly higher than that in the NRs at week 0. We included 850 K methylation chip sequencing, combining previous data of metagenomic sequencing, and metabolomic sequencing for multi-omics analysis to discuss the relationship between flora-metabolite-methylation in FMT. Finally, we found that hexanoic acid treatment can up-regulate the global methylation of peripheral blood mononuclear cells in SLE patients. Overall, our results delineate changes in methylation level after FMT treatment of SLE and reveal possible mechanisms of FMT treatment in terms of the recovery of abnormal hypomethylation.


Asunto(s)
Metilación de ADN , Lupus Eritematoso Sistémico , Humanos , Trasplante de Microbiota Fecal , Helicasa Inducida por Interferón IFIH1/genética , Leucocitos Mononucleares , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/terapia
4.
Curr Rheumatol Rep ; 25(6): 107-116, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37083877

RESUMEN

PURPOSE OF REVIEW: Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by various autoantibodies and multi-organ. Microbiota dysbiosis in the gut, skin, oral, and other surfaces has a significant impact on SLE development. This article summarizes relevant research and provides new microbiome-related strategies for exploring the mechanisms and treating patients with SLE. RECENT FINDINGS: SLE patients have disruptions in multiple microbiomes, with the gut microbiota (bacteria, viruses, and fungi) and their metabolites being the most thoroughly researched. This dysbiosis can promote SLE progression through mechanisms such as the leaky gut, molecular mimicry, and epigenetic regulation. Notwithstanding study constraints on the relationship between microbiota and SLE, specific interventions targeting the gut microbiota, such as probiotics, dietary management, and fecal microbiota transplantation, have emerged as promising SLE therapeutics.


Asunto(s)
Microbioma Gastrointestinal , Lupus Eritematoso Sistémico , Microbiota , Humanos , Disbiosis/microbiología , Epigénesis Genética , Microbioma Gastrointestinal/fisiología
5.
Front Med ; 16(5): 686-700, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36318354

RESUMEN

Systemic lupus erythematosus (SLE) is a complicated autoimmune disease affecting multiple systems and organs. It is highly heterogeneous, and it preferentially affects women at childbearing age, causing worldwide social burden. The pathogenesis of SLE mostly involves genetic predisposition, epigenetic dysregulation, overactivation of the immune system, and environment factors. Human microbiome, which is mostly composed of microbiota colonized in the gut, skin, and oral cavity, provides a natural microbiome barrier against environmental risks. The past decade of research has demonstrated a strong association between microbiota and metabolic diseases or gastrointestinal diseases. However, the role of microbiota in autoimmunity remains largely unknown until recently, when the technological and methodological progress facilitates further microbiota research in SLE. In this review, the latest research about the role and mechanisms of microbiota in SLE and the advances in the development of diagnostic and therapeutic strategies based on microbiota for SLE were summarized.


Asunto(s)
Microbioma Gastrointestinal , Lupus Eritematoso Sistémico , Microbiota , Humanos , Femenino , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/terapia , Autoinmunidad , Sistema Inmunológico
6.
J Exp Clin Cancer Res ; 41(1): 198, 2022 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-35689245

RESUMEN

BACKGROUND: NCAPD3 is one of the three non-SMC subunits of condensin II complex, which plays an important role in the chromosome condensation and segregation during mitosis. Notably, elevated levels of NCAPD3 are found in many somatic cancers. However, the clinical role, biological functions of NCAPD3 in cancers especially in colorectal cancer (CRC) and the underlying molecular mechanisms remain poorly elucidated. METHODS: Clinical CRC and adjacent normal tissues were used to confirm the expression of NCAPD3. The association of NCAPD3 expression with clinicopathological characteristics and patient outcomes were analyzed by using online database. In vivo subcutaneous tumor xenograft model, NCAPD3 gene knockout following azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced tumor mouse model, Co-IP, western blot, qRT-PCR, IHC, ChIP assays and cell functional assays were used to investigate the biological functions of NCAPD3 in CRC and the underlying molecular mechanisms. RESULTS: NCAPD3 was overexpressed in CRC tissues and positively correlated with poor prognosis of CRC patients. NCAPD3 knockout suppressed CRC development in AOM/DSS induced and xenograft mice models. Moreover, we found that NCAPD3 promoted aerobic glycolysis in CRC. Mechanistically, NCAPD3 up-regulated the level of c-Myc and interacted with c-Myc to recruit more c-Myc to the gene promoter of its downstream glycolytic regulators GLUT1, HK2, ENO1, PKM2 and LDHA, and finally enhanced cellular aerobic glycolysis. Also, NCAPD3 increased the level of E2F1 and interacted with E2F1 to recruit more E2F1 to the promoter regions of PDK1 and PDK3 genes, which resulted in the inhibition of PDH activity and TCA cycle. CONCLUSIONS: Our data demonstrated that NCAPD3 promoted glucose metabolism reprogramming and enhanced Warburg effect in colorectal tumorigenesis and CRC progression. These findings reveal a novel mechanism underlying NCAPD3 mediated CRC cell growth and provide new targets for CRC treatment.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorrectales , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Ratones
7.
Expert Opin Drug Discov ; 17(5): 489-500, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35287523

RESUMEN

INTRODUCTION: Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease with substantial phenotypic heterogeneity. Currently, our understanding of the pathogenesis is still limited, and as a result, specific and efficacious therapies are lacking. Various mouse models have been established to serve as powerful tools that will promote a better understanding of the disease and the ability to test novel drugs before clinical application. AREAS COVERED: The authors review the existing mouse models of SLE in terms of pathogenesis and manifestations, as well as their applications in drug discovery and development. The areas of focus include promising novel therapeutics that could benefit patients in the future and the contribution of mouse models used in preclinical studies. EXPERT OPINION: Given the diversity of SLE mouse models with different characteristics, researchers must select a suitable model based on the mechanism involved. The use of multiple models is needed for drug testing studies to evaluate drug efficacy on different genetic backgrounds and other mechanisms to provide a reference for clinical trials.


Asunto(s)
Lupus Eritematoso Sistémico , Animales , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , Ratones
8.
Cell Signal ; 92: 110265, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35085770

RESUMEN

NCAPD3 is one of the non-SMC regulatory subunits of Condensin II, which is mainly responsible for the condensation and segregation of chromosomes during mitosis. However, its role in cancer especially in prostate cancer (PCa) and the molecular mechanism have not been clearly elucidated. Here, we find that NCAPD3 is high-expression and up-regulates the levels of EZH2 and MALAT1 in PCa. In detail, high expression of NCAPD3 increases the levels of transcription factor STAT3 and E2F1 and recruits more STAT3 and E2F1 to the promoter of EZH2 gene and more STAT3 to the promoter of MALAT1 gene, and then results in the increasing expression of both EZH2 and MALAT1 in PCa cells. In vitro and in vivo functional characterization reveals that overexpression of NCAPD3 enhances the growth of PCa cells, while knockdown of NCAPD3 impairs the growth of PCa cells. Together, our data demonstrate that NCAPD3 is a tumor-promoting factor which enhances the progression of PCa by up-regulating EZH2 and MALAT1.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias de la Próstata , ARN Largo no Codificante/genética , Línea Celular Tumoral , Factor de Transcripción E2F1/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo
9.
Prostate ; 82(1): 26-40, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34591337

RESUMEN

BACKGROUND: Androgen receptor (AR) is an essential transcriptional factor that contributes to the development and progression of prostate cancer (PCa). NCAPD3 is a component of the condensin II complex and plays a critical role in cell mitosis by regulating chromosome condensation; however, the relationship between NCAPD3 and AR remains unknown. METHODS: Transcriptome sequencing assay is carried out to analyze the expression of the NCAP family in clinic samples. Chromatin immunoprecipitation (ChIP) sequencing, ChIP assay, and dual-luciferase assay are used to identify the androgen-responsive element in NCAPD3 enhancer. Immunohistochemistry, quantitative reverse transcription-polymerase chain reaction, and western-blot assay are employed to check the expression of genes in PCa tissues and in PCa cells. Confocal immunofluorescence microscopy analysis is used for identifying the regulation of AR on NCAPD3-mediated chromosome condensation. Colony formation, cell cycle assay, wound healing assay, and transwell experiments are used to explore the regulation of AR on the functions of NCAPD3. In vivo experiment is employed to identify in vitro experimental results. RESULTS: NCAPD3 is an androgen/AR axis-targeted gene and is involved in AR-induced PCa cell proliferation, migration, and invasion in vitro and in vivo. Androgen treatment and AR overexpression increase the expression of NCAPD3 in PCa cell lines. The canonical exist in the enhancer region of NCAPD3. Androgen/AR axis regulates NCAPD3-invovled chromosome condensation during cell mitosis. CONCLUSIONS: Our report demonstrated that NCAPD3 is an androgen-responsive gene and upregulated by androgen/AR axis and involved in AR-promoted progression of PCa, suggesting a potential role of NCAPD3 in the PCa development.


Asunto(s)
Proteínas de Ciclo Celular , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Adenosina Trifosfatasas/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Ensayos de Migración Celular/métodos , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Descubrimiento de Drogas , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Complejos Multiproteicos/metabolismo , Regulación hacia Arriba , Secuenciación del Exoma/métodos
10.
Pak J Med Sci ; 35(1): 236-240, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30881430

RESUMEN

OBJECTIVE: To discuss the influence of coronary artery lesion of elderly patients with coronary heart disease (CHD) on left ventricular remodeling. METHODS: Retrospective selection method was used to choose 80 elderly CHD patients who received coronary angiogram examination in Baoding First Central Hospital from January 2014 to February 2018 as the objects of study. According to coronary artery lesion, the patients were classified into single vessel lesion group (single vessel group) and multi-vessel lesion group (multi-vessel group, the number of lesion vessels≧2). Single vessel group included 60 patients, and multi-vessel group includes 20 patients. Intravascular unltrasound was applied to record coronary plaque property of all patients and transthoracic echocardiography was used to record left ventricular remodeling. Later correlation analysis was carried out. RESULTS: The proportion of calcified plaque and mixed plaque was higher than that of single vessel group, and the differences had statistical significance (P<0.05). Left ventricular end diastolic volume (LVEDV) and left ventricular end-systolic volume (LVESV) of multi-vessel group were higher than that of single vessel group, while left ventricular ejection fraction (LVEF) was lower than that of single vessel group. The differences had statistical significance (P<0.05). Linear correlation analysis showed coronary artery lesion was positively correlated with LVEF and calcified plaque (r=0.287, 0.371, P<0.05). Multiple linear regression analysis showed LVEF, calcified plaque and LDL-C were independent risk factors of multi-vessel coronary artery lesion of old CHD patients (P<0.05). CONCLUSION: The number of coronary artery lesions is significantly correlated with left ventricular remodeling, and can increase the proportion of calcified plaque and mixed plaque, thus leading to left ventricular remodeling abnormity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA