Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(5): e2316304121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261617

RESUMEN

The discovery that Africans were resistant to infection by Plasmodium vivax (P. vivax) led to the conclusion that P. vivax invasion relied on the P. vivax Duffy Binding Protein (PvDBP) interacting with the Duffy Antigen Receptor for Chemokines (DARC) expressed on erythrocytes. However, the recent reporting of P. vivax infections in DARC-negative Africans suggests that the parasite might use an alternate invasion pathway to infect DARC-negative reticulocytes. To identify the parasite ligands and erythrocyte receptors that enable P. vivax invasion of both DARC-positive and -negative erythrocytes, we expressed region II containing the Duffy Binding-Like (DBL) domain of P. vivax erythrocyte binding protein (PvEBP-RII) and verified that the DBL domain binds to both DARC-positive and -negative erythrocytes. Furthermore, an AVidity-based EXtracelluar Interaction Screening (AVEXIS) was used to identify the receptor for PvEBP among over 750 human cell surface receptor proteins, and this approach identified only Complement Receptor 1 (CR1, CD35, or C3b/C4b receptor) as a PvEBP receptor. CR1 is a well-known receptor for P. falciparum Reticulocyte binding protein Homology 4 (PfRh4) and is present on the surfaces of both reticulocytes and normocytes, but its expression decreases as erythrocytes age. Indeed, PvEBP-RII bound to a subpopulation of both reticulocytes and normocytes, and this binding was blocked by the addition of soluble CR1 recombinant protein, indicating that CR1 is the receptor of PvEBP. In addition, we found that the Long Homology Repeat A (LHR-A) subdomain of CR1 is the only subdomain responsible for mediating the interaction with PvEBP-RII.


Asunto(s)
Malaria Falciparum , Plasmodium vivax , Humanos , Receptores de Superficie Celular , Eritrocitos , Reticulocitos , Antígenos CD2 , Moléculas de Adhesión Celular
2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35210361

RESUMEN

5-methylcytosine (m5C) is an important epitranscriptomic modification involved in messenger RNA (mRNA) stability and translation efficiency in various biological processes. However, it remains unclear if m5C modification contributes to the dynamic regulation of the transcriptome during the developmental cycles of Plasmodium parasites. Here, we characterize the landscape of m5C mRNA modifications at single nucleotide resolution in the asexual replication stages and gametocyte sexual stages of rodent (Plasmodium yoelii) and human (Plasmodium falciparum) malaria parasites. While different representations of m5C-modified mRNAs are associated with the different stages, the abundance of the m5C marker is strikingly enhanced in the transcriptomes of gametocytes. Our results show that m5C modifications confer stability to the Plasmodium transcripts and that a Plasmodium ortholog of NSUN2 is a major mRNA m5C methyltransferase in malaria parasites. Upon knockout of P. yoelii nsun2 (pynsun2), marked reductions of m5C modification were observed in a panel of gametocytogenesis-associated transcripts. These reductions correlated with impaired gametocyte production in the knockout rodent malaria parasites. Restoration of the nsun2 gene in the knockout parasites rescued the gametocyte production phenotype as well as m5C modification of the gametocytogenesis-associated transcripts. Together with the mRNA m5C profiles for two species of Plasmodium, our findings demonstrate a major role for NSUN2-mediated m5C modifications in mRNA transcript stability and sexual differentiation in malaria parasites.


Asunto(s)
5-Metilcitosina/química , Plasmodium falciparum/metabolismo , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/metabolismo , Proteínas Protozoarias/metabolismo , ARN Mensajero/metabolismo , Células Germinativas , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium yoelii/genética , Transcriptoma
3.
Cancer Sci ; 115(1): 83-93, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37985391

RESUMEN

Autoantibodies (AAbs) in the blood of colorectal cancer (CRC) patients have been evaluated for tumor detection. However, it remains uncertain whether these AAbs are specific to tumor-associated antigens. In this study, we explored the IgG and IgM autoantibody repertoires in both the in situ tissue microenvironment and peripheral blood as potential tumor-specific biomarkers. We applied high-density protein arrays to profile AAbs in the tumor-infiltrating lymphocyte supernatants and corresponding serum from four patients with CRC, as well as in the serum of three noncancer controls. Our findings revealed that there were more reactive IgM AAbs than IgG in both the cell supernatant and corresponding serum, with a difference of approximately 3-5 times. Immunoglobulin G was predominant in the serum, while IgM was more abundant in the cell supernatant. We identified a range of AAbs present in both the supernatant and the corresponding serum, numbering between 432 and 780, with an average of 53.3% shared. Only 4.7% (n = 23) and 0.2% (n = 2) of reactive antigens for IgG and IgM AAbs, respectively, were specific to CRC. Ultimately, we compiled a list of 19 IgG AAb targets as potential tumor-specific AAb candidates. Autoantibodies against one of the top candidates, p15INK4b-related sequence/regulation of nuclear pre-mRNA domain-containing protein 1A (RPRD1A), were significantly elevated in 53 CRC patients compared to 119 controls (p < 0.0001). The project revealed that tissue-derived IgG AAbs, rather than IgM, are the primary source of tumor-specific AAbs in peripheral blood. It also identified potential tumor-specific AAbs that could be applied for noninvasive screening of CRC.


Asunto(s)
Autoanticuerpos , Neoplasias Colorrectales , Humanos , Biomarcadores de Tumor , Inmunoglobulina G , Inmunoglobulina M , Microambiente Tumoral , Proteínas Represoras , Proteínas de Ciclo Celular
4.
Nucleic Acids Res ; 50(6): 3413-3431, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35288749

RESUMEN

Heterochromatin-associated gene silencing controls multiple physiological processes in malaria parasites, however, little is known concerning the regulatory network and cis-acting sequences involved in the organization of heterochromatin and how they modulate heterochromatic gene expression. Based on systematic profiling of genome-wide occupancy of eighteen Apicomplexan AP2 transcription factors by ChIP-seq analysis, we identify and characterize eight heterochromatin-associated factors (PfAP2-HFs), which exhibit preferential enrichment within heterochromatic regions but with differential coverage profiles. Although these ApiAP2s target euchromatic gene loci via specific DNA motifs, they are likely integral components of heterochromatin independent of DNA motif recognition. Systematic knockout screenings of ApiAP2 factors coupled with RNA-seq transcriptomic profiling revealed three activators and three repressors of heterochromatic gene expression including four PfAP2-HFs. Notably, expression of virulence genes is either completely silenced or significantly reduced upon the depletion of PfAP2-HC. Integrated multi-omics analyses reveal autoregulation and feed-forward loops to be common features of the ApiAP2 regulatory network, in addition to the occurrence of dynamic interplay between local chromatin structure and ApiAP2s in transcriptional control. Collectively, this study provides a valuable resource describing the genome-wide landscape of the ApiAP2 family and insights into functional divergence and cooperation within this family during the blood-stage development of malaria parasites.


Asunto(s)
Malaria , Plasmodium falciparum , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Malaria/parasitología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-38818583

RESUMEN

Alcoholic liver disease (ALD) poses a significant health challenge, so comprehensive research efforts to improve our understanding and treatment strategies are needed. However, the development of effective treatments is hindered by the limitation of existing liver disease models. Liver organoids, characterized by their cellular complexity and three-dimensional (3D) tissue structure closely resembling the human liver, hold promise as ideal models for liver disease research. In this study, we use a meticulously designed protocol involving the differentiation of human induced pluripotent stem cells (hiPSCs) into liver organoids. This process incorporates a precise combination of cytokines and small molecule compounds within a 3D culture system to guide the differentiation process. Subsequently, these differentiated liver organoids are subject to ethanol treatment to induce ALD, thus establishing a disease model. A rigorous assessment through a series of experiments reveals that this model partially recapitulates key pathological features observed in clinical ALD, including cellular mitochondrial damage, elevated cellular reactive oxygen species (ROS) levels, fatty liver, and hepatocyte necrosis. In addition, this model offers potential use in screening drugs for ALD treatment. Overall, the liver organoid model of ALD, which is derived from hiPSC differentiation, has emerged as an invaluable platform for advancing our understanding and management of ALD in clinical settings.

6.
Proc Natl Acad Sci U S A ; 116(1): 255-260, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30584102

RESUMEN

Genetic manipulation remains a major obstacle for understanding the functional genomics of the deadliest malaria parasite Plasmodium falciparum Although the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeat/CRISPR-associated protein 9) system has been successfully applied to introduce permanent changes in the parasite genome, its use is still limited. Here we show that fusing different epigenetic effector domains to a Cas9 null mutant efficiently and specifically reprograms the expression of target genes in P. falciparum By precisely writing and erasing histone acetylation at the transcription start site regions of the invasion-related genes reticulocyte binding protein homolog 4 (rh4) and erythrocyte binding protein 175 (eba-175), respectively, we achieved significant activation of rh4 and repression of eba-175, leading to the switch of the parasite invasion pathways into human erythrocytes. By using the epigenetic knockdown system, we have also characterized the effects of PfSET1, previously identified as an essential gene, on expression of mainly trophozoite- and schizont-specific genes, and therefore regulation of the growth of the mature forms of P. falciparum This epigenetic CRISPR/dCas9 system provides a powerful approach for regulating gene expression at the transcriptional level in P. falciparum.


Asunto(s)
Sistemas CRISPR-Cas , Epigénesis Genética , Edición Génica/métodos , Plasmodium falciparum/genética , Proteína 9 Asociada a CRISPR/genética , Eritrocitos/parasitología , Técnicas de Silenciamiento del Gen , Genes Protozoarios/genética , Histona Acetiltransferasas/genética , Histona Desacetilasas/genética , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/fisiología , Proteínas Recombinantes
7.
Proc Natl Acad Sci U S A ; 115(5): 1063-1068, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339517

RESUMEN

Recent advances have identified a new paradigm for cerebral malaria pathogenesis in which endothelial protein C receptor (EPCR) is a major host receptor for sequestration of Plasmodium falciparum-infected erythrocytes (IEs) in the brain and other vital organs. The parasite adhesins that bind EPCR are members of the IE variant surface antigen family Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) containing specific adhesion domains called domain cassette (DC) 8 and DC13. The binding interaction site between PfEMP1 and EPCR has been mapped by biophysical and crystallography studies using recombinant proteins. However, studies examining the interaction of native PfEMP1 on the IE surface with EPCR are few. We aimed to study binding to EPCR by IEs expressing DC8 and DC13 PfEMP1 variants whose recombinant proteins have been used in key prior functional and structural studies. IE binding to EPCR immobilized on plastic and on human brain endothelial cells was examined in static and flow adhesion assays. Unexpectedly, we found that IEs expressing the DC13 PfEMP1 variant HB3var03 or IT4var07 did not bind to EPCR on plastic and the binding of these variants to brain endothelial cells was not dependent on EPCR. IEs expressing the DC8 variant IT4var19 did bind to EPCR, but this interaction was inhibited if normal human serum or plasma was present, raising the possibility that IE-EPCR interaction may be prevented by plasma components under physiological conditions. These data highlight a discrepancy in EPCR-binding activity between PfEMP1 recombinant proteins and IEs, and indicate the critical need for further research to understand the pathophysiological significance of the PfEMP1-EPCR interaction.


Asunto(s)
Eritrocitos/parasitología , Malaria Cerebral/parasitología , Malaria Falciparum/parasitología , Oligopéptidos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Adhesión Celular , Línea Celular , Receptor de Proteína C Endotelial/metabolismo , Epítopos/química , Humanos , Microcirculación , Peso Molecular , Unión Proteica , ARN Interferente Pequeño/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/metabolismo
8.
Proc Natl Acad Sci U S A ; 115(24): 6285-6290, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29844160

RESUMEN

Malaria control is threatened by a limited pipeline of effective pharmaceuticals against drug-resistant strains of Plasmodium falciparum Components of the mitochondrial electron transport chain (ETC) are attractive targets for drug development, owing to exploitable differences between the parasite and human ETC. Disruption of ETC function interferes with metabolic processes including de novo pyrimidine synthesis, essential for nucleic acid replication. We investigated the effects of ETC inhibitor selection on two distinct P. falciparum clones, Dd2 and 106/1. Compounds CK-2-68 and RYL-552, substituted quinolones reported to block P. falciparum NADH dehydrogenase 2 (PfNDH2; a type II NADH:quinone oxidoreductase), unexpectedly selected mutations at the quinol oxidation (Qo) pocket of P. falciparum cytochrome B (PfCytB). Selection experiments with atovaquone (ATQ) on 106/1 parasites yielded highly resistant PfCytB Y268S mutants seen in clinical infections that fail ATQ-proguanil treatment. In contrast, ATQ pressure on Dd2 yielded moderately resistant parasites carrying a PfCytB M133I or K272R mutation. Strikingly, all ATQ-selected mutants demonstrated little change or slight increase of sensitivity to CK-2-68 or RYL-552. Molecular docking studies demonstrated binding of all three ETC inhibitors to the Qo pocket of PfCytB, where Y268 forms strong van der Waals interactions with the hydroxynaphthoquinone ring of ATQ but not the quinolone ring of CK-2-68 or RYL-552. Our results suggest that combinations of suitable ETC inhibitors may be able to subvert or delay the development of P. falciparum drug resistance.


Asunto(s)
Citocromos b/genética , NADH Deshidrogenasa/antagonistas & inhibidores , Plasmodium falciparum/genética , Antimaláricos/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/genética , Malaria Falciparum/parasitología , Simulación del Acoplamiento Molecular/métodos , Mutación/genética , Plasmodium falciparum/efectos de los fármacos , Quinolonas/farmacología
9.
Proc Natl Acad Sci U S A ; 115(49): 12513-12518, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30455312

RESUMEN

Concerns about malaria parasite resistance to treatment with artemisinin drugs (ARTs) have grown with findings of prolonged parasite clearance t1/2s (>5 h) and their association with mutations in Plasmodium falciparum Kelch-propeller protein K13. Here, we describe a P. falciparum laboratory cross of K13 C580Y mutant with C580 wild-type parasites to investigate ART response phenotypes in vitro and in vivo. After genotyping >400 isolated progeny, we evaluated 20 recombinants in vitro: IC50 measurements of dihydroartemisinin were at similar low nanomolar levels for C580Y- and C580-type progeny (mean ratio, 1.00; 95% CI, 0.62-1.61), whereas, in a ring-stage survival assay, the C580Y-type progeny had 19.6-fold (95% CI, 9.76-39.2) higher average counts. In splenectomized Aotus monkeys treated with three daily doses of i.v. artesunate, t1/2 calculations by three different methods yielded mean differences of 0.01 h (95% CI, -3.66 to 3.67), 0.80 h (95% CI, -0.92 to 2.53), and 2.07 h (95% CI, 0.77-3.36) between C580Y and C580 infections. Incidences of recrudescence were 57% in C580Y (4 of 7) versus 70% in C580 (7 of 10) infections (-13% difference; 95% CI, -58% to 35%). Allelic substitution of C580 in a C580Y-containing progeny clone (76H10) yielded a transformant (76H10C580Rev) that, in an infected monkey, recrudesced regularly 13 times over 500 d. Frequent recrudescences of ART-treated P. falciparum infections occur with or without K13 mutations and emphasize the need for improved partner drugs to effectively eliminate the parasites that persist through the ART component of combination therapy.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Malaria Falciparum/parasitología , Plasmodium falciparum/efectos de los fármacos , Animales , Aotidae , Cruzamientos Genéticos , Resistencia a Medicamentos , Regulación de la Expresión Génica , Mutación , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
10.
J Med Virol ; 92(12): 3717-3725, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32558959

RESUMEN

Nasopharyngeal carcinoma (NPC), the most common head and neck cancer, is characterized by distinct geographic distribution and familial aggregation. Multiple risk factors, including host genetics, environmental factor, and EBV infection, have been linked to the development of NPC, particularly in the familial clustering cases. However, the cause of NPC endemicity remains enigmatic due possibly to the complicated interplay between these risk factors. Recently, positive Epstein-Barr virus (EBV) DNA loads at nasopharyngeal (NP) cavity has been found to reflect NPC development and applied in NPC screening. To examine whether the increased NP EBV loads could aggregate in the families and be affected by host genetics and environmental factor, EBV loads were obtained by 510 NP brushing samples from eligible unaffected individuals, who have two or more relatives affected with NPC, in 116 high-risk NPC families. The correlation of relative pairs was estimated using S.A.G.E. (version 6.4, 2016), and host heritability of NP EBV loads was calculated with variance component models using SOLAR (version 8.4.2, 2019). In result, significant correlations of EBV loads were observed between parent-offspring pairs and sibling-sibling pairs (P < .001), but not in distant kin relationship pairs. Interestingly, after excluding the shared environmental factor within families, host genetics contributes significantly to NP EBV loads with a heritability of 56.41% (P = 1.00 × 10-7 ), and its effect was slightly elevated (68.86%, P = 3.40 × 10-6 ) in families with more NPC cases (≥3). These findings indicate that additional host-genetic variants involved in the EBV local NP mucosal behavior may be especially important for the development of NPC.

11.
Acta Biochim Biophys Sin (Shanghai) ; 52(10): 1156-1165, 2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-33083831

RESUMEN

The central nervous system (CNS) diseases are still a major cause of morbidity and mortality throughout the world, which imposes heavy burden on the development of society. Ethionine is a non-proteinogenic amino acid having similar chemical structure and activity to that of methionine, with which it competes. Previous studies have confirmed that ethionine affects various cellular functions by inhibiting the biosynthesis of proteins, RNA, DNA, and phospholipids, or all of them. The relationship of ethionine with some CNS diseases, including neural tube defects, has been investigated recently. However, the detailed effects of ethionine on the nerve cell bioactivities and the underlying mechanisms have not been fully explored. Herein, we systematically investigated the influences of ethionine on the proliferation, differentiation, and apoptosis of neural stem cells (NSCs) and post-mitotic nerve cells. We demonstrated that ethionine inhibited cell viability by disrupting the balance between proliferation and apoptosis, prevented NSCs from differentiating into neurons and astrocytes, and blocked cell progression from G1 to S phase via reducing cyclin D1 function in nerve cells including NSCs, a mouse hippocampal neuron cell line (HT-22), and a mouse brain neuroma cell line (Neuro-2a). We speculated that the inhibitory effect of ethionine on cell viability and differentiation are associated with increased reactive oxygen species production. Our results also supported the concept that ethionine may be an underlying cause of abnormal folate metabolism-induced CNS diseases. Our findings may provide important direction for the application of abnormal folate metabolism-induced CNS diseases in future NSC-based therapies.


Asunto(s)
Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Etionina/farmacología , Células-Madre Neurales/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Astrocitos/metabolismo , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Enfermedades del Sistema Nervioso Central/etiología , Enfermedades del Sistema Nervioso Central/metabolismo , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Proteína X Asociada a bcl-2/metabolismo
12.
Environ Toxicol ; 35(9): 961-970, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32255272

RESUMEN

Benzo[a]pyrene (B[a]P) is recognized as a neurotoxic pollutant to mammals, which could impair learning and memory function. Although there is some evidence to suggest that N-methyl-d-aspartate receptor (NMDAR), a glutamate receptor and ion channel protein in nerve cells, is involved into the B[a]P induced neurotoxicity, the exact molecular mechanisms remain to be elucidated, particularly the effects of B[a]P on the NMDAR downstream signaling transduction pathways. In the present study, we examined the neurotoxicity of sub-chronic administrated B[a]P on male Sprague-Dawley rats. Our data suggested that B[a]P exposure caused significant deficits in learning and memory function and the impairment of hippocampal LTP in rats. Further mechanistic studies indicate that B[a]P-induced learning and memory deficits are associated with the inhibition of NMDAR NR1 subunit transcription and protein phosphorylation. More importantly, the inactivation of CaMK II/PKC/PKA-ERK-CREB signaling pathways in hippocampus was detected at both the 2.5 and 6.25 mg/kg B[a]P-treated groups, indicating that multiple targets in NMDAR and downstream signaling pathways are involved in the B[a]P-induced neurotoxicity.


Asunto(s)
Benzo(a)pireno/toxicidad , Región CA1 Hipocampal/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Proteína Quinasa C/metabolismo , Receptores de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Región CA1 Hipocampal/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
13.
J Infect Dis ; 219(3): 400-409, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30307559

RESUMEN

Background: Oral Epstein-Barr virus (EBV) status reflects host EBV activity and potentially links to EBV-associated diseases, however, factors influencing oral EBV loads or reactivation, such as environmental exposures or host factors, are not fully understood. Methods: A 2-stage, multicenter, cross-sectional study of 6558 subjects from 21 administrative cities of southern China and 3 populations from representative geographical areas in China (referred to as the south, north, and northeastern populations) was performed. The relationships between demographical factors and environmental exposures to EBV loads were analyzed by logistic regression models. Results: Current smoking, with a dose-response effect, was found to be strongly associated with higher oral EBV loads in the pooled data, with an odds ratio of 1.58 (95% confidence interval, 1.39-1.79), as well as in each of the separate populations. The odds ratio increased to 3.06 when current smokers in southern China were compared to never smokers in northern China. Additionally, higher oral EBV loads tended to be detected in older participants, male participants, and participants in southern China. Conclusions: This study provided evidence linking the effect of host-environmental factors, particularly smoking, to oral EBV activity. It could strengthen our understanding of the possible causal roles of EBV-related diseases, which may help to prevent or mitigate EBV-associated diseases.


Asunto(s)
ADN Viral , Demografía , Exposición a Riesgos Ambientales , Herpesvirus Humano 4/genética , Boca/virología , Adulto , Anciano , Anciano de 80 o más Años , China , Estudios Transversales , Infecciones por Virus de Epstein-Barr/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Población , Análisis de Regresión , Fumar , Carga Viral , Adulto Joven
14.
BMC Infect Dis ; 19(1): 999, 2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775660

RESUMEN

BACKGROUND: Recent studies have shown that CD103 is an important marker for tissue-resident memory T cells (TRM) which plays an important role in anti-infection. However, the role of CD103+ TRM was not elucidated in the progress of S. japonicum infection induced disease. METHODS: 6-8 weeks old C57BL/6 mice were infected by S. japonicum. Mice were sacrificed and the lungs were removed 5-6 weeks after infection. Immunofluorescent staining and Q-PCR were performed to identify the expression of CD103 molecule. Single cellular populations were made, percentages of CD103 on both CD4+ and CD8+ T lymphocytes were dynamical observed by flow cytometry (FCM). Moreover, the expression of memory T cells related molecules CD69 and CD62L, T cell function associated molecules CD107a, IFN-γ, IL-4, IL-9, and IL-10 were compared between CD103+ CD4+ and CD8+ T cells by FCM. RESULTS: CD103+ cells were emerged in the lung of both naive and S. japonicum infected mice. Both the percentage and the absolute numbers of pulmonary CD4+ and CD8+ cells were increased after S. japonicum infection (P < 0.05). The percentage of CD103+ cells in CD8+ T cells decreased significantly at the early stage of S. japonicum infection (P < 0.05). Increased CD69, decreased CD62L and CD107a expressions were detected on both CD4+ and CD8+ CD103+ T cells in the lungs of infected mice (P < 0.05). Compared to CD8+ CD103+ T cells, CD4+ CD103+ T cells from infected mice expressed higher level of CD69 and lower level CD62L molecules (P < 0.05). Moreover, higher percentage of IL-4+, IL-9+ and IL-10+ cells on CD4+ CD103+ pulmonary T cells was found in infected mice (P < 0.05). Significantly increased IL-4 and IL-9, and decreased IFN-γ expressing cells were detected in CD8+CD103+ cells of infected mice (P < 0.05). CONCLUSIONS: CD103-expressing pulmonary CD4+ and CD8+ T cells play important roles in mediating S. japonicum infection induced granulomatous inflammation in the lung.


Asunto(s)
Antígenos CD/genética , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Schistosoma japonicum , Esquistosomiasis Japónica/metabolismo , Animales , Biomarcadores/metabolismo , Citocinas/metabolismo , Femenino , Expresión Génica/inmunología , Memoria Inmunológica , Pulmón/metabolismo , Pulmón/parasitología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Esquistosomiasis Japónica/microbiología
15.
J Immunol ; 198(12): 4716-4727, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28476935

RESUMEN

Myeloid-derived suppressor cells (MDSCs), a heterogeneous group of immune cells from the myeloid lineage, play an important part in suppression of host immune responses during many pathologic conditions, including cancer and infectious diseases. Thus, understanding the functional diversity of these cells as well as the underlying mechanisms is crucial for the development of disease control strategies. The role of MDSCs during Schistosoma japonicum infection, however, is not clear, and there is a lack of systematic study so far. In this study, we provide strong evidence that the soluble egg Ag (SEA) and schistosome worm Ag (SWA) of S. japonicum enhance the accumulation of MDSCs. Ag-induced MDSCs have more potent suppressive effects on T cell responses than do control MDSCs in both in vivo S. japonicum infection and in vitro SEA- and SWA-treated mouse bone marrow cells experiments. Interestingly, the enhanced suppressive activity of MDSCs by Ag administration was coupled with a dramatic induction of the NADPH oxidase subunits gp91phox and p47phox and was dependent on the production of reactive oxygen species. Moreover, mechanistic studies revealed that the Ag effects are mediated by JAK/STAT3 signaling. Inhibition of STAT3 phosphorylation by the JAK inhibitor JSI-124 almost completely abolished the Ag effects on the MDSCs. In summary, this study sheds new light on the immune modulatory role of SEA and SWA and demonstrates that the expansion of MDSCs may be an important element of a cellular network regulating immune responses during S. japonicum infection.


Asunto(s)
Quinasas Janus/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Factor de Transcripción STAT3/metabolismo , Esquistosomiasis Japónica/metabolismo , Transducción de Señal , Animales , Proliferación Celular , Regulación de la Expresión Génica , Ratones , Células Supresoras de Origen Mieloide/inmunología , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Schistosoma japonicum/inmunología , Esquistosomiasis Japónica/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Triterpenos/farmacología
16.
Nature ; 499(7457): 223-7, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23823717

RESUMEN

The variant antigen Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1), which is expressed on the surface of P. falciparum-infected red blood cells, is a critical virulence factor for malaria. Each parasite has 60 antigenically distinct var genes that each code for a different PfEMP1 protein. During infection the clonal parasite population expresses only one gene at a time before switching to the expression of a new variant antigen as an immune-evasion mechanism to avoid the host antibody response. The mechanism by which 59 of the 60 var genes are silenced remains largely unknown. Here we show that knocking out the P. falciparum variant-silencing SET gene (here termed PfSETvs), which encodes an orthologue of Drosophila melanogaster ASH1 and controls histone H3 lysine 36 trimethylation (H3K36me3) on var genes, results in the transcription of virtually all var genes in the single parasite nuclei and their expression as proteins on the surface of individual infected red blood cells. PfSETvs-dependent H3K36me3 is present along the entire gene body, including the transcription start site, to silence var genes. With low occupancy of PfSETvs at both the transcription start site of var genes and the intronic promoter, expression of var genes coincides with transcription of their corresponding antisense long noncoding RNA. These results uncover a previously unknown role of PfSETvs-dependent H3K36me3 in silencing var genes in P. falciparum that might provide a general mechanism by which orthologues of PfSETvs repress gene expression in other eukaryotes. PfSETvs knockout parasites expressing all PfEMP1 proteins may also be applied to the development of a malaria vaccine.


Asunto(s)
Silenciador del Gen , Histonas/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Factores de Virulencia/genética , Proteínas de Unión al ADN , Proteínas de Drosophila , Eritrocitos/citología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Genes Protozoarios/genética , Histonas/química , Intrones/genética , Lisina/metabolismo , Vacunas contra la Malaria/genética , Metilación , Plasmodium falciparum/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Protozoarias/genética , ARN Largo no Codificante/genética , Factores de Transcripción , Sitio de Iniciación de la Transcripción , Virulencia/genética
17.
Proc Natl Acad Sci U S A ; 113(22): 6271-6, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27190089

RESUMEN

The ability of the malaria parasite Plasmodium vivax to invade erythrocytes is dependent on the expression of the Duffy blood group antigen on erythrocytes. Consequently, Africans who are null for the Duffy antigen are not susceptible to P. vivax infections. Recently, P. vivax infections in Duffy-null Africans have been documented, raising the possibility that P. vivax, a virulent pathogen in other parts of the world, may expand malarial disease in Africa. P. vivax binds the Duffy blood group antigen through its Duffy-binding protein 1 (DBP1). To determine if mutations in DBP1 resulted in the ability of P. vivax to bind Duffy-null erythrocytes, we analyzed P. vivax parasites obtained from two Duffy-null individuals living in Ethiopia where Duffy-null and -positive Africans live side-by-side. We determined that, although the DBP1s from these parasites contained unique sequences, they failed to bind Duffy-null erythrocytes, indicating that mutations in DBP1 did not account for the ability of P. vivax to infect Duffy-null Africans. However, an unusual DNA expansion of DBP1 (three and eight copies) in the two Duffy-null P. vivax infections suggests that an expansion of DBP1 may have been selected to allow low-affinity binding to another receptor on Duffy-null erythrocytes. Indeed, we show that Salvador (Sal) I P. vivax infects Squirrel monkeys independently of DBP1 binding to Squirrel monkey erythrocytes. We conclude that P. vivax Sal I and perhaps P. vivax in Duffy-null patients may have adapted to use new ligand-receptor pairs for invasion.


Asunto(s)
Antígenos de Protozoos/genética , Eritrocitos/parasitología , Malaria Vivax/parasitología , Mutación/genética , Plasmodium vivax/genética , Proteínas Protozoarias/genética , Receptores de Superficie Celular/genética , África/epidemiología , Animales , Variaciones en el Número de Copia de ADN , Sistema del Grupo Sanguíneo Duffy/genética , Eritrocitos/patología , Humanos , Malaria Vivax/epidemiología , Malaria Vivax/genética , Malaria Vivax/patología , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Saimiri
18.
Chin J Cancer Res ; 31(3): 463-470, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31354215

RESUMEN

OBJECTIVE: Tumor microenvironment, especially the host immune system, plays a pivotal role in tumor initiation and progression. Profiling of immune signature within tumor might uncover biomarkers for targeted therapies and clinical outcomes. However, systematic analysis of immune-related genes in gastric cancer (GC) has not been reported. METHODS: Expressions of a total of 718 immune-related genes were generated in 372 stomach adenocarcinoma (STAD) patients from The Cancer Genome Atlas (TCGA) database using RNA-sequencing data. Integrated bioinformatics analyses were performed to identify prognostic factors as well. RESULTS: Survival analyses revealed 73 genes, which were significantly associated with patient's overall survival (OS). Taken together with clinicopathological parameters, we established a predictive model, containing 10 immune-related genes, which were NRP1, C6, CXCR4, LBP, PNMA1, TLR5, ITGA6, MICB, PBK and TNFRSF18, with powerful efficiency in distinguishing satisfactory or poor survival of STAD patients. Moreover, the top 3 ranked prognostic genes, NRP1, TGFß2 and MFGE8, were also significantly associated with patient's OS by an independent validation achieved from Kaplan-Meier plotter database. CONCLUSIONS: We profiled prognostic immune signature and established prognostic predictive model for GC, which could reflect immune disorders within tumor microenvironment, and also may provide novel predictive and therapeutic targets for GC patients in the near future.

19.
Malar J ; 17(1): 60, 2018 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-29394891

RESUMEN

BACKGROUND: Four Plasmodium falciparum genetic crosses (HB3×3D7, HB3×Dd2, 7G8×GB4, and 803×GB4) have produced sets of recombinant progeny that are widely used for malaria research, including investigations of anti-malarial drug resistance. It is critical to maintain the progeny free from cross-contamination. Microsatellite polymorphisms can be used to validate parasite identity. RESULTS: A set of 12 markers was developed that differentiates the parents of the four P. falciparum crosses. This typing set identified distinguishing patterns of inheritance (fingerprints) in segregant collections of 15 progeny clones from HB3×3D7, 32 from HB3×Dd2, 33 from 7G8×GB4, and 81 from 803×GB4. Stronger amplification was observed with shorter relative to longer alleles of individual microsatellites. In experiments with mixed parental DNAs, electropherograms showed that signals of cross-contamination can be missed when minor peaks less than 1/4 or 1/3 the height of the major peak are disregarded by threshold settings commonly used for population studies. CONCLUSIONS: Microsatellite typing is an effective method to check the identity of P. falciparum lines and detect parasite cross-contamination in cultures; however, care must be taken not to ignore minor peaks that can be overlooked. The 12 microsatellite markers presented here provide a rapid and efficient means to distinguish the segregants of laboratory crosses. Fingerprint patterns from these markers are useful to maintain the integrity of diverse parasite lines in and between research laboratories.


Asunto(s)
ADN Protozoario/genética , Genotipo , Repeticiones de Microsatélite/genética , Plasmodium falciparum/genética , Marcadores Genéticos/genética , Técnicas de Genotipaje , Humanos , Malaria Falciparum/parasitología , Tipificación Molecular
20.
Occup Environ Med ; 75(11): 807-813, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30217924

RESUMEN

OBJECTIVES: The interaction between genetic, epigenetic inheritance and environmental factors determines susceptibility to hypertension. Previous epidemiology studies have shown that coke oven workers who are frequently exposed to various occupational hazards have remarkable increase in the risk for hypertension. Among many genetic variants identified in hypertension, heat shock protein 70 (HSP70) was found to play important roles in the pathogenesis of hypertension and associated diseases. We therefore explore the possible role of HSP70 polymorphisms and their interaction with occupational environment in hypertension risk. METHODS: We carried out a case-control study among 367 coke oven workers in northwest China, focused on three common HSP70 polymorphisms (HSP70-1 G190C, HSP70-2 A1267G and HSP70-hom T2437C), and evaluated the association of HSP70 gene polymorphisms with work sites for high risk of hypertension. RESULTS: The results indicated that HSP70-1 GC and CC genotype had 2.73-fold and 4.26-fold increased relative risk (95% CI 1.33 to 5.55 and 1.17 to 15.53), respectively, comparing with HSP70-1 GG genotype. HSP70-2 AG and GG conferred a 47% and 36% reduced risk (95% CI 0.23 to 0.99 and 0.14 to 0.92) comparing with HSP70-2 AA genotype. Further analysis of the interaction of HSP70 polymorphisms with occupational environment indicated a strong positive interaction between HSP70 genotype (HSP70-1 GC+CC, HSP70-2 AA and HSP70-hom TC+CC) and oven top workplace. CONCLUSIONS: Collectively, these data indicate that HSP70 polymorphisms interact with occupational hazards might increase the risk of hypertension in coke oven workers.


Asunto(s)
Coque/toxicidad , Predisposición Genética a la Enfermedad , Proteínas HSP70 de Choque Térmico/genética , Hipertensión/genética , Exposición Profesional/efectos adversos , Polimorfismo de Nucleótido Simple/genética , Adulto , Estudios de Casos y Controles , China , Interacción Gen-Ambiente , Genotipo , Calor/efectos adversos , Humanos , Hipertensión/etiología , Industrias , Masculino , Análisis de Regresión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA