Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Respir Res ; 22(1): 274, 2021 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-34696775

RESUMEN

BACKGROUND: Dysbiosis of the gut microbiome is involved in the pathogenesis of various diseases, but the contribution of gut microbes to the progression of chronic obstructive pulmonary disease (COPD) is still poorly understood. METHODS: We carried out 16S rRNA gene sequencing and short-chain fatty acid analyses in stool samples from a cohort of 73 healthy controls, 67 patients with COPD of GOLD stages I and II severity, and 32 patients with COPD of GOLD stages III and IV severity. Fecal microbiota from the three groups were then inoculated into recipient mice for a total of 14 times in 28 days to induce pulmonary changes. Furthermore, fecal microbiota from the three groups were inoculated into mice exposed to smoke from biomass fuel to induce COPD-like changes. RESULTS: We observed that the gut microbiome of COPD patients varied from that of healthy controls and was characterized by a distinct overall microbial diversity and composition, a Prevotella-dominated gut enterotype and lower levels of short-chain fatty acids. After 28 days of fecal transplantation from COPD patients, recipient mice exhibited elevated lung inflammation. Moreover, when mice were under both fecal transplantation and biomass fuel smoke exposure for a total of 20 weeks, accelerated declines in lung function, severe emphysematous changes, airway remodeling and mucus hypersecretion were observed. CONCLUSION: These data demonstrate that altered gut microbiota in COPD patients is associated with disease progression in mice model.


Asunto(s)
Bacterias/crecimiento & desarrollo , Microbioma Gastrointestinal , Intestinos/microbiología , Pulmón/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Anciano , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Bacterias/genética , Bacterias/metabolismo , Estudios de Casos y Controles , China , Estudios Transversales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Disbiosis , Ácidos Grasos Volátiles/metabolismo , Trasplante de Microbiota Fecal , Heces/química , Heces/microbiología , Femenino , Humanos , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Ribotipificación
2.
Respir Res ; 21(1): 271, 2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-33076910

RESUMEN

BACKGROUND: The role of the microbiota in the pathogenesis of chronic obstructive pulmonary disease (COPD) following exposure to ambient particulate matter (PM) is largely unknown. METHODS: Fifty-four male Sprague-Dawley rats were exposed to clean air, biomass fuel (BMF), or motor vehicle exhaust (MVE) for 4, 12, and 24 weeks. We performed pulmonary inflammation evaluation, morphometric measurements, and lung function analysis in rat lung at three different times points during exposure. Lung and gut microbial composition was assessed by 16S rRNA pyrosequencing. Serum lipopolysaccharide levels were measured and short-chain fatty acids in colon contents were quantified. RESULTS: After a 24-week PM exposure, rats exhibited pulmonary inflammation and pathological changes characteristic of COPD. The control and PM exposure (BMF and MVE) groups showed similar microbial diversity and composition in rat lung. However, the gut microbiota after 24 weeks PM exposure was characterized by decreased microbial richness and diversity, distinct overall microbial composition, lower levels of short-chain fatty acids, and higher serum lipopolysaccharide. CONCLUSION: Chronic exposure to ambient particulate matter induces gut microbial dysbiosis and metabolite shifts in a rat model of chronic obstructive pulmonary disease.


Asunto(s)
Disbiosis/inducido químicamente , Microbioma Gastrointestinal/efectos de los fármacos , Exposición por Inhalación/efectos adversos , Pulmón/efectos de los fármacos , Material Particulado/toxicidad , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Animales , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Disbiosis/sangre , Disbiosis/fisiopatología , Microbioma Gastrointestinal/fisiología , Pulmón/fisiopatología , Masculino , Material Particulado/administración & dosificación , Enfermedad Pulmonar Obstructiva Crónica/sangre , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Ratas , Ratas Sprague-Dawley
3.
Mol Cancer ; 16(1): 20, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28137278

RESUMEN

BACKGROUND: Colorectal cancer remains one of the most common malignant tumors worldwide. Colorectal cancer initiating cells (CCICs) are a small subpopulation responsible for malignant behaviors of colorectal cancer. Aberrant activation of the Wnt pathways regulates the self-renewal of CCIC. However, the underlying mechanism(s) remain poorly understood. METHODS: Via retroviral library screening, we identified Nuclear Receptor-Interacting Protein 2 (NRIP2) as a novel interactor of the Wnt pathway from enriched colorectal cancer colosphere cells. The expression levels of NRIP2 and retinoic acid-related orphan receptor ß (RORß) were further examined by FISH, qRT-PCR, IHC and Western blot. NRIP2 overexpressed and knockdown colorectal cancer cells were produced to study the role of NRIP2 in Wnt pathway. We also verified the binding between NRIP2 and RORß and investigated the effect of RORß on CCICs both in vitro and in vivo. Genechip-scanning speculated downstream target HBP1. Western blot, ChIP and luciferase reporter were carried to investigate the interaction between NRIP2, RORß, and HBP1. RESULTS: NRIP2 was significantly up-regulated in CCICs from both cell lines and primary colorectal cancer tissues. Reinforced expression of NRIP2 increased Wnt activity, while silencing of NRIP2 attenuated Wnt activity. The transcription factor RORß was a key target through which NRIP2 regulated Wnt pathway activity. RORß was a transcriptional enhancer of inhibitor HBP1 of the Wnt pathway. NRIP2 prevented RORß to bind with downstream HBP1 promoter regions and reduced the transcription of HBP1. This, in turn, attenuated the HBP1-dependent inhibition of TCF4-mediated transcription. CONCLUSIONS: NRIP2 is a novel interactor of the Wnt pathway in colorectal cancer initiating cells. interactions between NRIP2, RORß, and HBP1 mediate a new mechanism for CCIC self-renewal via the Wnt activity.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/genética , Proteínas del Grupo de Alta Movilidad/genética , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/genética , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Proteínas Represoras/genética , Regulación hacia Arriba , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HT29 , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Ratones , Trasplante de Neoplasias , Proteínas del Tejido Nervioso/metabolismo , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Represoras/metabolismo , Vía de Señalización Wnt
4.
Life Sci ; 351: 122802, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38857656

RESUMEN

Adenosine nucleotide translocases (ANTs) are a family of proteins abundant in the inner mitochondrial membrane, primarily responsible for shuttling ADP and ATP across the mitochondrial membrane. Additionally, ANTs are key players in balancing mitochondrial energy metabolism and regulating cell death. ANT2 isoform, highly expressed in undifferentiated and proliferating cells, is implicated in the development and drug resistance of various tumors. We conduct a detailed analysis of the potential mechanisms by which ANT2 may influence tumorigenesis and drug resistance. Notably, the significance of ANT2 extends beyond oncology, with roles in non-tumor cell processes including blood cell development, gastrointestinal motility, airway hydration, nonalcoholic fatty liver disease, obesity, chronic kidney disease, and myocardial development, making it a promising therapeutic target for multiple pathologies. To better understand the molecular mechanisms of ANT2, this review summarizes the structural properties, expression patterns, and basic functions of the ANT2 protein. In particular, we review and analyze the controversy surrounding ANT2, focusing on its role in transporting ADP/ATP across the inner mitochondrial membrane, its involvement in the composition of the mitochondrial permeability transition pore, and its participation in apoptosis.


Asunto(s)
Translocador 2 del Nucleótido Adenina , Humanos , Animales , Translocador 2 del Nucleótido Adenina/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Apoptosis , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Translocasas Mitocondriales de ADP y ATP/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Adenosina Trifosfato/metabolismo
5.
Int Immunopharmacol ; 117: 109917, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36822087

RESUMEN

BACKGROUND: During organ transplantation, pharmacologic drugs targeting T cell activation signal to inhibit T cell-mediated allo-rejection are insufficient and not durable to suppress chronic rejection. Recent advances highlight an exhausted or dysfunctional status of T cells, which favor transplant acceptance. METHODS: The models of MHC-mismatched (BALB/c to C57BL/6 or USP25 KO mice) heterotopic heart transplantation and skin transplantation were utilized to evaluate the regulatory effects of ubiquitin-specific protease 25(USP25) deficiency in vivo. The consequences of USP25 deficiency on murine T-cell proliferation, activation, cytokine secretion, mixed lymphocyte reaction (MLR) and energy metabolism were investigated in vitro. The signaling pathway of T cells in knock out mice was detected by Western blotting and Co-IP. RESULTS: We found T cells were dysfunctional inUSP25KO mice. Due to T cell dysfunction, skin and heart graft had a longer survival. In these dysfunctional T cells, mitochondria number and cristae condensation were decreased. Impaired mitochondrial mass and function favored to allo-graft acceptance. Furthermore, USP25 interacted with ATP5A and ATP5B to promote their stability. CONCLUSIONS: Our data suggest that USP25 is a potential target to induce T cell dysfunction and allo-graft tolerance. And USP25 mediated mitochondrial homeostasis may contribute to reverse T cell exhaustion or dysfunction in tumor and chronic infection.


Asunto(s)
Trasplante de Corazón , Trasplante de Órganos , Ratones , Animales , Dinámicas Mitocondriales , Ratones Endogámicos C57BL , Tolerancia al Trasplante , Linfocitos T , Ratones Noqueados , Ratones Endogámicos BALB C , Rechazo de Injerto/patología , Supervivencia de Injerto
6.
Front Med (Lausanne) ; 9: 838738, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35308516

RESUMEN

ABO blood group antibodies have not been generated or are at low titer during early infancy. Therefore, in theory, ABO-incompatible kidney transplantation (ABOi KT) may be successfully achieved in small infants without any pre-transplant treatment. We report here the first ABO-incompatible deceased donor kidney transplantation (ABOi DDKT) in an infant. The recipient infant was ABO blood group O, and the donor group A. The recipient was diagnosed with a Wilms tumor gene 1 (WT1) mutation and had received peritoneal dialysis for 4 months prior to transplant. At 7 months and 27 days of age, the infant underwent bilateral native nephrectomy and single-kidney transplantation from a 3-year-old brain-dead donor. No pre- or post-transplantation antibody removal treatment was performed, since the recipient's anti-iso-hemagglutinin-A Ig-M/G antibody titers were both low (1:2) before transplantation and have remained at low levels or undetectable to date. At 11 months post-transplant, the recipient is at home, thriving, with normal development and graft function. This outcome suggests that ABOi DDKT without antibody removal preparatory treatment is feasible in small infants, providing a new option for kidney transplantation in this age range.

7.
Theranostics ; 10(2): 516-536, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31903135

RESUMEN

Background: The leading cause of poor prognosis in colorectal cancer (CRC) is the presence of colorectal cancer-initiating cells (CCICs). The interplay between the tumor microenvironment (TME) and CRC cells induces reacquisition of initiating cell characteristics, but the underlying mechanisms remain elusive. Methods: Candidate molecules were screened by global differential cDNA expression profiles of CCICs, which were enriched from patient-derived tumor xenograft models. Luciferase reporters and chromatin immunoprecipitation assays were used to explore the mechanism of TME factors regulating the transcription of ANKRD22. The effects of Ankyrin repeat domain-containing protein 22 (ANKRD22) on energy metabolism were monitored by extracellular flux and 13C-based metabolic flux analysis. Mass spectrometry was used to identify the interacting partners of ANKRD22. Morphological changes of CCICs overexpressing ANKRD22 were observed by electron microscopy. The effects of ANKRD22 on mitochondrial lipid metabolism were analyzed by lipidomics. Results: We identified a novel nucleus-encoded mitochondrial membrane protein, ANKRD22, which was upregulated in CCICs. We found that ANKRD22 was induced by the p38/MAX pathway activated by different TME stimuli. As a key transcription factor, MAX promoted the transcription of ANKRD22. Expression of ANKRD22 promoted glycolysis associated with a decrease in ATP/ADP and an increase in AMP/ATP levels, which were related to its interaction with pyruvate dehydrogenase kinase isoform 1 (PDK1) and multiple subunits of ATP synthase. Further, in CCICs, ANKRD22 cooperated with the lipid transport protein, Extended Synaptotagmin-1 (E-Syt1), to transport excess lipids into mitochondria and reduced the number of mitochondria in an autophagy-independent manner, thus meeting the metabolic requirements of CCICs. Conclusion: ANKRD22 induced by TME promotes the metabolic reprogramming of CRC cells. Our study has identified ANKRD22/E-Syt1 as a potential target for eradicating CCICs.


Asunto(s)
Autofagia , Reprogramación Celular , Neoplasias Colorrectales/patología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Lipidómica/métodos , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas Mitocondriales/genética , Esferoides Celulares , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncol Lett ; 18(4): 4106-4113, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31516611

RESUMEN

Prostate cancer is a common malignant tumor in elderly men. As a novel metabolic-reprogramming molecule, the role of ankyrin repeat domain 22 (ANKRD22) in the tumorigenesis and progression of prostate cancer remains unknown. In the present study, mouse monoclonal antibodies against human ANKRD22 were prepared using recombinant ANKRD22 from prokaryotic expression and validated. Subsequently, these antibodies were used to evaluate ANKRD22 levels via immunohistochemical staining in prostate cancer tissues. Finally, the association between ANKRD22 levels and prostate cancer progression was analyzed in 636 samples of prostate cancer using The Cancer Genome Atlas (TCGA) database. A total of four anti-ANKRD22 monoclonal antibodies were generated and validated, which could be effectively blocked by recombinant ANKRD22 protein. Using these antibodies for immunohistochemical staining, ANKRD22 was detected in prostate cancer cells in both the cytoplasm and nucleus. Bioinformatics analysis demonstrated that the mRNA level of ANKRD22 was inversely associated with prostate cancer stage (P<0.05) and Gleason score (P<0.01) in TCGA database. Patients with higher ANKRD22 mRNA levels exhibited longer disease-free survival following radical prostatectomy. These findings suggest that ANKRD22 may negatively regulate the progression of prostate cancer. The prepared ANKRD22 antibodies with high specificity provide a powerful tool in ANKRD22 research.

9.
Int J Mol Med ; 39(1): 9-20, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27909729

RESUMEN

Colorectal cancer stem cells (CCSCs) represent a small fraction of the colorectal cancer cell population that possess self-renewal and multi-lineage differentiation potential and drive tumorigenicity. Self-renewal is essential for the malignant biological behaviors of colorectal cancer stem cells. While the self-renewal molecular mechanisms of colorectal cancer stem cells are not yet fully understood, the aberrant activation of signaling pathways, such as Wnt, Notch, transforming growth factor-ß (TGF-ß)/bone morphogenetic protein (BMP) and Hedgehog-Gli (HH-GLI), specific roles mediated by cell surface markers and micro-environmental factors are involved in the regulation of self-renewal. The elucidation of the molecular mechanisms behind self-renewal may lead to the development of novel targeted interventions for the treatment of colorectal cancer.


Asunto(s)
Autorrenovación de las Células , Neoplasias Colorrectales/patología , Células Madre Neoplásicas/patología , Animales , Humanos , Inflamación/patología , Transducción de Señal , Microambiente Tumoral
10.
Sci Rep ; 5: 12230, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26178819

RESUMEN

Pancreatic cancer is one of the more common cancers with a poor prognosis. Some varieties of cancer are related to virus infection. As a virus-induced protein, APOBEC3G (A3G) presents extensive anti-virus ability, but the role of A3G in pancreatic cancer was previously unknown. The expression of A3G in pancreatic cancer was examined using TaqMan real-time qPCR, immunohistochemical and immunofluorescent staining. Subsequently, the role of A3G in pancreatic cancer was evaluated in vivo using the tumor xenograft model. Anoikis was detected by colony formation assay and flow cytometry in vitro. The Akt kinase activity and target protein PTEN were examined by co-immunoprecipitation and immunoblot. The virus-induced protein A3G was significantly up-regulated in pancreatic cancer, and the up-regulation of A3G promoted xenograft tumor formation. A3G inactivated PTEN by binding to the C2 tensin-type and PDZ domains, thereby inducing anoikis resistance through Akt activation. Our results demonstrate that the up-regulation of A3G in pancreatic cancer cells induces anoikis resistance, and they provide novel insight into the mechanism by which A3G affects the malignant behavior of pancreatic cancer cells.


Asunto(s)
Anoicis/fisiología , Citidina Desaminasa/fisiología , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Desaminasa APOBEC-3G , Animales , Sitios de Unión , Línea Celular Tumoral , Activación Enzimática , Humanos , Ratones , Ratones Desnudos , Fosfohidrolasa PTEN/fisiología , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA