Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.270
Filtrar
1.
PLoS One ; 19(5): e0302913, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38728358

RESUMEN

In the fight against antimicrobial resistance, host defense peptides (HDPs) are increasingly referred to as promising molecules for the design of new antimicrobial agents. In terms of their future clinical use, particularly small, synthetic HDPs offer several advantages, based on which their application as feed additives has aroused great interest in the poultry sector. However, given their complex mechanism of action and the limited data about the cellular effects in production animals, their investigation is of great importance in these species. The present study aimed to examine the immunomodulatory activity of the synthetic HDP Pap12-6 (PAP) solely and in inflammatory environments evoked by lipoteichoic acid (LTA) and polyinosinic-polycytidylic acid (Poly I:C), in a primary chicken hepatocyte-non-parenchymal cell co-culture. Based on the investigation of the extracellular lactate dehydrogenase (LDH) activity, PAP seemed to exert no cytotoxicity on hepatic cells, suggesting its safe application. Moreover, PAP was able to influence the immune response, reflected by the decreased production of interleukin (IL)-6, IL-8, and "regulated on activation, normal T cell expressed and secreted"(RANTES), as well as the reduced IL-6/IL-10 ratio in Poly I:C-induced inflammation. PAP also diminished the levels of extracellular H2O2 and nuclear factor erythroid 2-related factor 2 (Nrf2) when applied together with Poly I:C and in both inflammatory conditions, respectively. Consequently, PAP appeared to display potent immunomodulatory activity, preferring to act towards the cellular anti-inflammatory and antioxidant processes. These findings confirm that PAP might be a promising alternative for designing novel antimicrobial immunomodulatory agents for chickens, thereby contributing to the reduction of the use of conventional antibiotics.


Asunto(s)
Pollos , Hepatocitos , Lipopolisacáridos , Poli I-C , Animales , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/metabolismo , Poli I-C/farmacología , Lipopolisacáridos/farmacología , Factores Inmunológicos/farmacología , Ácidos Teicoicos/farmacología , Células Cultivadas , Agentes Inmunomoduladores/farmacología , Agentes Inmunomoduladores/química , Técnicas de Cocultivo , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Citocinas/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología
2.
J Math Biol ; 88(6): 75, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689137

RESUMEN

The aim of this paper is to develop and investigate a novel mathematical model of the dynamical behaviors of chronic hepatitis B virus infection. The model includes exposed infected hepatocytes, intracellular HBV DNA-containing capsids, uses a general incidence function for viral infection covering a variety of special cases available in the literature, and describes the interaction of cytotoxic T lymphocytes that kill the infected hepatocytes and the magnitude of B-cells that send antibody immune defense to neutralize free virions. Further, one time delay is incorporated to account for actual capsids production. The other time delays are used to account for maturation of capsids and free viruses. We start with the analysis of the proposed model by establishing the local and global existence, uniqueness, non-negativity and boundedness of solutions. After defined the threshold parameters, we discuss the stability properties of all possible steady state constants by using the crafty Lyapunov functionals, the LaSalle's invariance principle and linearization methods. The impacts of the three time delays on the HBV infection transmission are discussed through local and global sensitivity analysis of the basic reproduction number and of the classes of infected states. Finally, an application is provided and numerical simulations are performed to illustrate and interpret the theoretical results obtained. It is suggested that, a good strategy to eradicate or to control HBV infection within a host should concentrate on any drugs that may prolong the values of the three delays.


Asunto(s)
Inmunidad Adaptativa , Cápside , Simulación por Computador , Virus de la Hepatitis B , Hepatitis B Crónica , Hepatocitos , Conceptos Matemáticos , Hepatocitos/inmunología , Hepatocitos/virología , Virus de la Hepatitis B/inmunología , Humanos , Cápside/inmunología , Inmunidad Adaptativa/inmunología , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Hepatitis B Crónica/transmisión , Modelos Inmunológicos , Linfocitos T Citotóxicos/inmunología , Número Básico de Reproducción/estadística & datos numéricos , Linfocitos B/inmunología , ADN Viral/inmunología , Modelos Biológicos
3.
Arch Virol ; 169(5): 112, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683226

RESUMEN

Previously, we reported a neutralizing monoclonal antibody, A8A11, raised against a novel conserved epitope within the hepatitis C virus (HCV) E2 protein, that could significantly reduce HCV replication. Here, we report the nucleotide sequence of A8A11 and demonstrate the efficacy of a single-chain variable fragment (scFv) protein that mimics the antibody, inhibits the binding of an HCV virus-like particle to hepatocytes, and reduces viral RNA replication in a cell culture system. More importantly, scFv A8A11 was found to effectively restrict the increase of viral RNA levels in the serum of HCV-infected chimeric mice harbouring human hepatocytes. These results suggest a promising approach to neutralizing-antibody-based therapeutic interventions against HCV infection.


Asunto(s)
Epítopos , Hepacivirus , Hepatocitos , Anticuerpos de Cadena Única , Proteínas del Envoltorio Viral , Internalización del Virus , Hepacivirus/inmunología , Hepacivirus/genética , Hepacivirus/fisiología , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/genética , Hepatocitos/virología , Hepatocitos/inmunología , Animales , Humanos , Epítopos/inmunología , Ratones , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Hepatitis C/virología , Hepatitis C/inmunología , Anticuerpos Neutralizantes/inmunología , Replicación Viral , Anticuerpos Monoclonales/inmunología
4.
Nat Immunol ; 25(5): 755-763, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38641718

RESUMEN

T cell infiltration into tumors is a favorable prognostic feature, but most solid tumors lack productive T cell responses. Mechanisms that coordinate T cell exclusion are incompletely understood. Here we identify hepatocyte activation via interleukin-6/STAT3 and secretion of serum amyloid A (SAA) proteins 1 and 2 as important regulators of T cell surveillance of extrahepatic tumors. Loss of STAT3 in hepatocytes or SAA remodeled the tumor microenvironment with infiltration by CD8+ T cells, while interleukin-6 overexpression in hepatocytes and SAA signaling via Toll-like receptor 2 reduced the number of intratumoral dendritic cells and, in doing so, inhibited T cell tumor infiltration. Genetic ablation of SAA enhanced survival after tumor resection in a T cell-dependent manner. Likewise, in individuals with pancreatic ductal adenocarcinoma, long-term survivors after surgery demonstrated lower serum SAA levels than short-term survivors. Taken together, these data define a fundamental link between liver and tumor immunobiology wherein hepatocytes govern productive T cell surveillance in cancer.


Asunto(s)
Linfocitos T CD8-positivos , Hepatocitos , Interleucina-6 , Factor de Transcripción STAT3 , Proteína Amiloide A Sérica , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/genética , Hepatocitos/metabolismo , Hepatocitos/inmunología , Animales , Humanos , Ratones , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Microambiente Tumoral/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Escape del Tumor , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Línea Celular Tumoral
5.
Science ; 384(6691): eabo7027, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38574142

RESUMEN

Macrophages are functionally heterogeneous cells essential for apoptotic cell clearance. Apoptotic cells are defined by homogeneous characteristics, ignoring their original cell lineage identity. We found that in an interleukin-4 (IL-4)-enriched environment, the sensing of apoptotic neutrophils by macrophages triggered their tissue remodeling signature. Engulfment of apoptotic hepatocytes promoted a tolerogenic phenotype, whereas phagocytosis of T cells had little effect on IL-4-induced gene expression. In a mouse model of parasite-induced pathology, the transfer of macrophages conditioned with IL-4 and apoptotic neutrophils promoted parasitic egg clearance. Knockout of phagocytic receptors required for the uptake of apoptotic neutrophils and partially T cells, but not hepatocytes, exacerbated helminth infection. These findings suggest that the identity of apoptotic cells may contribute to the development of distinct IL-4-driven immune programs in macrophages.


Asunto(s)
Apoptosis , Interleucina-4 , Macrófagos , Fagocitosis , Esquistosomiasis mansoni , Animales , Ratones , Apoptosis/inmunología , Hepatocitos/inmunología , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/inmunología , Ratones Noqueados , Neutrófilos/inmunología , Fagocitosis/inmunología , Esquistosomiasis mansoni/genética , Esquistosomiasis mansoni/inmunología , Modelos Animales de Enfermedad
6.
Antiviral Res ; 226: 105896, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679167

RESUMEN

Immune tolerance to the hepatitis B virus (HBV) is crucial for developing chronic hepatitis B, and the HBV surface antigen (HBsAg) produced and secreted in high amounts is regarded as a key contributor. HBsAg is expressed in HBV-infected hepatocytes and those carrying an HBV integration. Whether either HBsAg secretion or the high antigen amount expressed in the liver determines its immunomodulatory properties, however, remains unclear. We, therefore, developed a novel HBV animal model that allowed us to study the role of secreted HBsAg. We introduced a previously described HBs mutation, C65S, abolishing HBsAg secretion into a replication-competent 1.3-overlength HBV genome and used adeno-associated virus vectors to deliver it to the mouse liver. The AAV-HBV established a carrier state of wildtype and C65S mutant HBV, respectively. We investigated antiviral B- and T-cell immunity in the HBV-carrier mice after therapeutic vaccination. Moreover, we compared the effect of a lacking HBsAg secretion with that of an antiviral siRNA. While missing HBsAg secretion allowed for higher levels of detectable anti-HBs antibodies after therapeutic vaccination, it did neither affect antiviral T-cell responses nor intrahepatic HBV gene expression, irrespective of the starting level. A treatment with HBV siRNA restricting viral antigen expression within hepatocytes, however, improved the antiviral efficacy of therapeutic vaccination, irrespective of the ability of HBV to secrete HBsAg. Our data indicate that clearing HBsAg from blood cannot significantly impact HBV persistence or T-cell immunity. This indicates that a restriction of hepatic viral antigen expression will be required to break HBV immunotolerance.


Asunto(s)
Modelos Animales de Enfermedad , Antígenos de Superficie de la Hepatitis B , Virus de la Hepatitis B , Linfocitos T , Animales , Antígenos de Superficie de la Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/genética , Ratones , Linfocitos T/inmunología , Hígado/inmunología , Hígado/virología , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Hepatitis B/inmunología , Hepatitis B/virología , Mutación , Ratones Endogámicos C57BL , Dependovirus/genética , Dependovirus/inmunología , Anticuerpos contra la Hepatitis B/sangre , Anticuerpos contra la Hepatitis B/inmunología , Hepatocitos/virología , Hepatocitos/inmunología , Humanos
7.
J Virol ; 98(3): e0192123, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38319104

RESUMEN

Hepatitis C virus (HCV) infection progresses to chronicity in the majority of infected individuals. Its high intra-host genetic variability enables HCV to evade the continuous selection pressure exerted by the host, contributing to persistent infection. Utilizing a cell culture-adapted HCV population (p100pop) which exhibits increased replicative capacity in various liver cell lines, this study investigated virus and host determinants that underlie enhanced viral fitness. Characterization of a panel of molecular p100 clones revealed that cell culture adaptive mutations optimize a range of virus-host interactions, resulting in expanded cell tropism, altered dependence on the cellular co-factor micro-RNA 122 and increased rates of virus spread. On the host side, comparative transcriptional profiling of hepatoma cells infected either with p100pop or its progenitor virus revealed that enhanced replicative fitness correlated with activation of endoplasmic reticulum stress signaling and the unfolded protein response. In contrast, infection of primary human hepatocytes with p100pop led to a mild attenuation of virion production which correlated with a greater induction of cell-intrinsic antiviral defense responses. In summary, long-term passage experiments in cells where selective pressure from innate immunity is lacking improves multiple virus-host interactions, enhancing HCV replicative fitness. However, this study further indicates that HCV has evolved to replicate at low levels in primary human hepatocytes to minimize innate immune activation, highlighting that an optimal balance between replicative fitness and innate immune induction is key to establish persistence. IMPORTANCE: Hepatitis C virus (HCV) infection remains a global health burden with 58 million people currently chronically infected. However, the detailed molecular mechanisms that underly persistence are incompletely defined. We utilized a long-term cell culture-adapted HCV, exhibiting enhanced replicative fitness in different human liver cell lines, in order to identify molecular principles by which HCV optimizes its replication fitness. Our experimental data revealed that cell culture adaptive mutations confer changes in the host response and usage of various host factors. The latter allows functional flexibility at different stages of the viral replication cycle. However, increased replicative fitness resulted in an increased activation of the innate immune system, which likely poses boundary for functional variation in authentic hepatocytes, explaining the observed attenuation of the adapted virus population in primary hepatocytes.


Asunto(s)
Aptitud Genética , Hepacivirus , Hepatocitos , Interacciones Microbiota-Huesped , Inmunidad Innata , Mutación , Humanos , Células Cultivadas , Estrés del Retículo Endoplásmico , Aptitud Genética/genética , Aptitud Genética/inmunología , Hepacivirus/genética , Hepacivirus/crecimiento & desarrollo , Hepacivirus/inmunología , Hepacivirus/fisiología , Hepatitis C/inmunología , Hepatitis C/virología , Hepatocitos/inmunología , Hepatocitos/virología , Interacciones Microbiota-Huesped/inmunología , MicroARNs/metabolismo , Pase Seriado , Respuesta de Proteína Desplegada , Tropismo Viral , Virión/crecimiento & desarrollo , Virión/metabolismo , Replicación Viral/genética , Replicación Viral/inmunología
8.
Nature ; 619(7971): 819-827, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37438530

RESUMEN

Understanding protective immunity to COVID-19 facilitates preparedness for future pandemics and combats new SARS-CoV-2 variants emerging in the human population. Neutralizing antibodies have been widely studied; however, on the basis of large-scale exome sequencing of protected versus severely ill patients with COVID-19, local cell-autonomous defence is also crucial1-4. Here we identify phospholipid scramblase 1 (PLSCR1) as a potent cell-autonomous restriction factor against live SARS-CoV-2 infection in parallel genome-wide CRISPR-Cas9 screens of human lung epithelia and hepatocytes before and after stimulation with interferon-γ (IFNγ). IFNγ-induced PLSCR1 not only restricted SARS-CoV-2 USA-WA1/2020, but was also effective against the Delta B.1.617.2 and Omicron BA.1 lineages. Its robust activity extended to other highly pathogenic coronaviruses, was functionally conserved in bats and mice, and interfered with the uptake of SARS-CoV-2 in both the endocytic and the TMPRSS2-dependent fusion routes. Whole-cell 4Pi single-molecule switching nanoscopy together with bipartite nano-reporter assays found that PLSCR1 directly targeted SARS-CoV-2-containing vesicles to prevent spike-mediated fusion and viral escape. A PLSCR1 C-terminal ß-barrel domain-but not lipid scramblase activity-was essential for this fusogenic blockade. Our mechanistic studies, together with reports that COVID-associated PLSCR1 mutations are found in some susceptible people3,4, identify an anti-coronavirus protein that interferes at a late entry step before viral RNA is released into the host-cell cytosol.


Asunto(s)
COVID-19 , Proteínas de Transferencia de Fosfolípidos , SARS-CoV-2 , Animales , Humanos , Ratones , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Quirópteros , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/prevención & control , COVID-19/virología , Secuenciación del Exoma , Hepatocitos/inmunología , Hepatocitos/metabolismo , Interferón gamma/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Fusión de Membrana , Proteínas de Transferencia de Fosfolípidos/química , Proteínas de Transferencia de Fosfolípidos/genética , Proteínas de Transferencia de Fosfolípidos/inmunología , Proteínas de Transferencia de Fosfolípidos/metabolismo , SARS-CoV-2/clasificación , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Internalización del Virus
9.
Front Immunol ; 14: 1140201, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36936935

RESUMEN

Background: Liver zonation is a unique phenomenon in which the liver exhibits distinct functions among hepatocytes along the radial axis of the lobule. This phenomenon can cause the sectionalized initiation of several liver diseases, including hepatocellular carcinoma (HCC). However, few studies have explored the zonation features of HCC. Methods: Four single-cell RNA sequencing datasets were used to identify hepatocyte-specific zonation markers. Integrative analysis was then performed with a training RNA-seq cohort (616 HCC samples) and an external validating microarray cohort (285 HCC samples) from the International Cancer Genome Consortium, The Cancer Genome Atlas, Gene Expression Omnibus, and EMBL's European Bioinformatics Institute for clustering using non-negative matrix factorization consensus clustering based on zonation genes. Afterward, we evaluated the prognostic value, clinical characteristics, transcriptome and mutation features, immune infiltration, and immunotherapy response of the HCC subclasses. Results: A total of 94 human hepatocyte-specific zonation markers (39 central markers and 55 portal markers) were identified for the first time. Subsequently, three subgroups of HCC, namely Cluster1, Cluster2, and Cluster3 were identified. Cluster1 exhibited a non-zonational-like signature with the worst prognosis. Cluster2 was intensively associated with a central-like signature and exhibited low immune infiltration and sensitivity toward immune blockade therapy. Cluster3 was intensively correlated with a portal-like signature with the best prognosis. Finally, we identified candidate therapeutic targets and agents for Cluster1 HCC samples. Conclusion: The current study established a novel HCC classification based on liver zonation signature. By classifying HCC into three clusters with non-zonational-like (Cluster1), central-like (Cluster2), and portal-like (Cluster3) features, this study provided new perspectives on the heterogeneity of HCC and shed new light on delivering precision medicine for HCC patients.


Asunto(s)
Biomarcadores , Carcinoma Hepatocelular , Neoplasias Hepáticas , Hígado , Fenotipo , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Carcinoma Hepatocelular/clasificación , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/terapia , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Transcriptoma , Mutación , Inmunoterapia , Neoplasias Hepáticas/clasificación , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Análisis de Expresión Génica de una Sola Célula , Análisis de Secuencia de ARN , Conjuntos de Datos como Asunto , Reproducibilidad de los Resultados , Estudios de Cohortes , Medicina de Precisión , Pronóstico , Terapia Molecular Dirigida , Algoritmos , Humanos , Animales , Ratones
10.
Curr Opin Immunol ; 80: 102267, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36462263

RESUMEN

The human liver mediates whole-body metabolism, systemic inflammation and responses to hepatotropic pathogens. Hepatocytes, the most abundant cell type of the liver, have critical roles in each of these activities. The regulation of metabolic pathways, such as glucose metabolism, lipid biosynthesis and oxidation, influences whole-organism functionality. However, the immune potential of the liver in general and hepatocytes in particular is also determined by metabolic ability. The major shifts in cellular metabolism required to drive activity in immune cells are now well-described. Given the unique functions of hepatocytes in systemic metabolism and inflammation, and their ability to mediate local antiviral innate immunity, the metabolic shifts required to facilitate these activities are likely to be complex and challenging to define. In this review, we explore what is known about the complex metabolic rewiring required for hepatocytes to respond appropriately to viral infection. We also discuss how viruses can manipulate hepatocyte metabolism to facilitate infection.


Asunto(s)
Hepatocitos , Inmunidad Innata , Virosis , Humanos , Hepatocitos/inmunología , Inflamación/metabolismo , Hígado , Virosis/inmunología
11.
Nature ; 611(7936): 563-569, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36352220

RESUMEN

Malaria infection involves an obligatory, yet clinically silent liver stage1,2. Hepatocytes operate in repeating units termed lobules, exhibiting heterogeneous gene expression patterns along the lobule axis3, but the effects of hepatocyte zonation on parasite development at the molecular level remain unknown. Here we combine single-cell RNA sequencing4 and single-molecule transcript imaging5 to characterize the host and parasite temporal expression programmes in a zonally controlled manner for the rodent malaria parasite Plasmodium berghei ANKA. We identify differences in parasite gene expression in distinct zones, including potentially co-adaptive programmes related to iron and fatty acid metabolism. We find that parasites develop more rapidly in the pericentral lobule zones and identify a subpopulation of periportally biased hepatocytes that harbour abortive infections, reduced levels of Plasmodium transcripts and parasitophorous vacuole breakdown. These 'abortive hepatocytes', which appear predominantly with high parasite inoculum, upregulate immune recruitment and key signalling programmes. Our study provides a resource for understanding the liver stage of Plasmodium infection at high spatial resolution and highlights the heterogeneous behaviour of both the parasite and the host hepatocyte.


Asunto(s)
Regulación de la Expresión Génica , Hepatocitos , Hígado , Malaria , Parásitos , Plasmodium berghei , Análisis de la Célula Individual , Animales , Hepatocitos/citología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/parasitología , Hígado/anatomía & histología , Hígado/citología , Hígado/inmunología , Hígado/parasitología , Malaria/genética , Malaria/inmunología , Malaria/parasitología , Parásitos/genética , Parásitos/inmunología , Parásitos/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Plasmodium berghei/metabolismo , Imagen Individual de Molécula , Análisis de Secuencia de ARN , Hierro/metabolismo , Ácidos Grasos/metabolismo , Transcripción Genética , Genes Protozoarios/genética , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología
12.
J Virol ; 96(7): e0199521, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35297672

RESUMEN

C-type lectin domain-containing proteins (CTLDcps) shape host responses to pathogens and infectious disease outcomes. Previously, we identified the murine CTLDcp Cd302 as restriction factor, limiting hepatitis C virus (HCV) infection of murine hepatocytes. In this study, we investigated in detail the human orthologue's ability to restrict HCV infection in human liver cells. CD302 overexpression in Huh-7.5 cells potently inhibited infection of diverse HCV chimeras representing seven genotypes. Transcriptional profiling revealed abundant CD302 mRNA expression in human hepatocytes, the natural cellular target of HCV. Knockdown of endogenously expressed CD302 modestly enhanced HCV infection of Huh-7.5 cells and primary human hepatocytes. Functional analysis of naturally occurring CD302 transcript variants and engineered CD302 mutants showed that the C-type lectin-like domain (CTLD) is essential for HCV restriction, whereas the cytoplasmic domain (CPD) is dispensable. Coding single nucleotide polymorphisms occurring in human populations and mapping to different domains of CD302 did not influence the capacity of CD302 to restrict HCV. Assessment of the anti-HCV phenotype at different life cycle stages indicated that CD302 preferentially targets the viral entry step. In contrast to the murine orthologue, overexpression of human CD302 did not modulate downstream expression of nuclear receptor-controlled genes. Ectopic CD302 expression restricted infection of liver tropic hepatitis E virus (HEV), while it did not affect infection rates of two respiratory viruses, including respiratory syncytial virus (RSV) and the alpha coronavirus HVCoV-229E. Together, these findings suggest that CD302 contributes to liver cell-intrinsic defense against HCV and might mediate broader antiviral defenses against additional hepatotropic viruses. IMPORTANCE The liver represents an immunoprivileged organ characterized by enhanced resistance to immune responses. However, the importance of liver cell-endogenous, noncytolytic innate immune responses in pathogen control is not well defined. Although the role of myeloid cell-expressed CTLDcps in host responses to viruses has been characterized in detail, we have little information about their potential functions in the liver and their relevance for immune responses in this organ. Human hepatocytes endogenously express the CTLDcp CD302. Here, we provide evidence that CD302 limits HCV infection of human liver cells, likely by inhibiting a viral cell entry step. We confirm that the dominant liver-expressed transcript variant, as well as naturally occurring coding variants of CD302, maintain the capacity to restrict HCV. We further show that the CTLD of the protein is critical for the anti-HCV activity and that overexpressed CD302 limits HEV infection. Thus, CD302 likely contributes to human liver-intrinsic antiviral defenses.


Asunto(s)
Hepacivirus , Hepatitis C , Lectinas Tipo C , Receptores de Superficie Celular , Antivirales/metabolismo , Hepacivirus/fisiología , Hepatitis C/inmunología , Hepatocitos/inmunología , Hepatocitos/virología , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Replicación Viral
13.
Front Immunol ; 13: 795463, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35197971

RESUMEN

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral 'TRM-like' cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM-like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.


Asunto(s)
Vacunas contra la Malaria/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos , Hepatocitos/inmunología , Humanos , Inmunidad Celular , Memoria Inmunológica/inmunología , Hígado/inmunología , Malaria/inmunología , Plasmodium/inmunología , Esporozoítos/inmunología , Transcriptoma , Vacunación
14.
Microbiol Spectr ; 10(1): e0132821, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35171034

RESUMEN

Chronic infection of hepatitis B virus (HBV) remains a major health burden worldwide. While the immune response has been recognized to play crucial roles in HBV pathogenesis, the direct cytopathic effects of HBV infection and replication on host hepatocytes and the HBV-host interactions are only partially defined due to limited culture systems. Here, based on our recently developed 5 chemical-cultured primary human hepatocytes (5C-PHHs) model that supports long-term HBV infection, we performed multiplexed quantitative analysis of temporal changes of host proteome and transcriptome on PHHs infected by HBV for up to 4 weeks. We showed that metabolic-, complement-, cytoskeleton-, mitochondrial-, and oxidation-related pathways were modulated at transcriptional or posttranscriptional levels during long-term HBV infection, which led to cytopathic effects and could be partially rescued by early, rather than late, nucleot(s)ide analog (NA) administration and could be significantly relieved by blocking viral antigens with RNA interference (RNAi). Overexpression screening of the dysregulated proteins identified a series of host factors that may contribute to pro- or anti-HBV responses of the infected hepatocytes. In conclusion, our results suggest that long-term HBV infection in primary human hepatocytes leads to cytopathic effects through remodeling the proteome and transcriptome and early antiviral treatment may reduce the extent of such effects, indicating a role of virological factors in HBV pathogenesis and a potential benefit of early administration of antiviral treatment. IMPORTANCE Global temporal quantitative proteomic and transcriptomic analysis using long-term hepatitis B virus (HBV)-infected primary human hepatocytes uncovered extensive remodeling of the host proteome and transcriptome and revealed cytopathic effects of long-term viral replication. Metabolic-, complement-, cytoskeleton-, mitochondrial-, and oxidation-related pathways were modulated at transcriptional or posttranscriptional levels, which could be partially rescued by early, rather than late, NA therapy and could be relieved by blocking viral antigens with RNAi. Overexpression screening identified a series of pro- or anti-HBV host factors. These data have deepened the understanding of the mechanisms of viral pathogenesis and HBV-host interactions in hepatocytes, with implications for therapeutic intervention.


Asunto(s)
Antivirales/farmacología , Efecto Citopatogénico Viral , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/fisiología , Hepatitis B/tratamiento farmacológico , Hepatocitos/virología , Técnicas de Cultivo de Célula , Guanina/análogos & derivados , Guanina/farmacología , Hepatitis B/genética , Hepatitis B/inmunología , Hepatitis B/virología , Virus de la Hepatitis B/genética , Hepatocitos/inmunología , Humanos , Modelos Biológicos , Transcriptoma/efectos de los fármacos , Replicación Viral
15.
Signal Transduct Target Ther ; 7(1): 23, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35078968
16.
Nat Commun ; 13(1): 105, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013224

RESUMEN

Zika virus (ZIKV) infection can be associated with neurological pathologies, such as microcephaly in newborns and Guillain-Barre syndrome in adults. Effective therapeutics are currently not available. As such, a comprehensive understanding of virus-host interactions may guide the development of medications for ZIKV. Here we report a human genome-wide overexpression screen to identify host factors that regulate ZIKV infection and find TMEM120A as a ZIKV restriction factor. TMEM120A overexpression significantly inhibits ZIKV replication, while TMEM120A knockdown increases ZIKV infection in cell lines. Moreover, Tmem120a knockout in mice facilitates ZIKV infection in primary mouse embryonic fibroblasts (MEF) cells. Mechanistically, the antiviral activity of TMEM120A is dependent on STING, as TMEM120A interacts with STING, promotes the translocation of STING from the endoplasmic reticulum (ER) to ER-Golgi intermediate compartment (ERGIC) and enhances the phosphorylation of downstream TBK1 and IRF3, resulting in the expression of multiple antiviral cytokines and interferon-stimulated genes. In summary, our gain-of-function screening identifies TMEM120A as a key activator of the antiviral signaling of STING.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Canales Iónicos/genética , Proteínas de la Membrana/genética , Infección por el Virus Zika/genética , Virus Zika/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Línea Celular Tumoral , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/virología , Femenino , Regulación de la Expresión Génica , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Aparato de Golgi/virología , Hepatocitos/inmunología , Hepatocitos/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Interferón beta/genética , Interferón beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Canales Iónicos/deficiencia , Canales Iónicos/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Virus Zika/crecimiento & desarrollo , Virus Zika/patogenicidad , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
17.
Alcohol Clin Exp Res ; 46(1): 40-51, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34773268

RESUMEN

BACKGROUND AND AIMS: Approximately 3.5% of the global population is chronically infected with Hepatitis B Virus (HBV), which puts them at high risk of end-stage liver disease, with the risk of persistent infection potentiated by alcohol consumption. However, the mechanisms underlying the effects of alcohol on HBV persistence remain unclear. Here, we aimed to establish in vivo/ex vivo evidence that alcohol suppresses HBV peptides-major histocompatibility complex (MHC) class I antigen display on primary human hepatocytes (PHH), which diminishes the recognition and clearance of HBV-infected hepatocytes by cytotoxic T-lymphocytes (CTLs). METHODS: We used fumarylacetoacetate hydrolase (Fah)-/-, Rag2-/-, common cytokine receptor gamma chain knock-out (FRG-KO) humanized mice transplanted with human leukocyte antigen-A2 (HLA-A2)-positive hepatocytes. The mice were HBV-infected and fed control and alcohol diets. Isolated hepatocytes were exposed ex vivo to HBV 18-27-HLA-A2-restricted CTLs to quantify cytotoxicity. For mechanistic studies, we measured proteasome activities, unfolded protein response (UPR), and endoplasmic reticulum (ER) stress in hepatocytes from HBV-infected humanized mouse livers. RESULTS AND CONCLUSIONS: We found that alcohol feeding attenuated HBV core 18-27-HLA-A2 complex presentation on infected hepatocytes due to the suppression of proteasome function and ER stress induction, which diminished both the processing of HBV peptides and trafficking of HBV-MHC class I complexes to the hepatocyte surface. This alcohol-mediated decrease in MHC class I-restricted antigen presentation of the CTL epitope on target hepatocytes reduced the CTL-specific elimination of infected cells, potentially leading to HBV-infection persistence, which promotes end-stage liver disease outcomes.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Etanol/farmacología , Virus de la Hepatitis B/inmunología , Hepatitis B/inmunología , Hepatocitos/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Enfermedad Hepática en Estado Terminal/virología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Antígeno HLA-A2/análisis , Hepatocitos/trasplante , Hepatocitos/virología , Xenoinjertos , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Ratones , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/fisiología , Respuesta de Proteína Desplegada/genética
18.
Life Sci ; 288: 120180, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34843736

RESUMEN

Alcoholic liver injury is a liver cell dysfunction disease caused by long-term or excessive alcohol consumption. Inhibiting the production of inflammatory factors is an important way to alleviate liver injury. Interleukin-9 (IL-9) is one of the members of IL-2Rγc family. It has multiple biological functions. Previous studies have shown that IL-9 is a cytokine that is closely related to inflammatory disease, allergic diseases, autoimmune diseases, and parasitic infections. However, no systematic studies have been performed to address the role of IL-9 in ALI. This project aims to investigate the effects of IL-9 on macrophage-related inflammatory response and hepatocyte apoptosis in alcohol-induced liver injury by injecting adeno-associated virus (AAV9) into tail vein. In the ALI model group, western blot and ELISA assays demonstrated that the expression of IL-9 was reduced. Overexpression of IL-9 relieved the injury and reduced the serum levels of IL-6, TNF-α in EtOH-induced ALI mouse model. Moreover, by using western blot, it was indicated that IL-9 can inhibit the expression of pro-apoptotic protein, such as cleaved caspase 3 and Bax. In vitro, mouse recombinant protein IL-9 inhibited the expression of IL-6, TNF-α in EtOH-induced RAW264.7 cells. Moreover, flow cytometry and western blot results displayed that macrophage-derived IL-9 inhibited hepatocyte apoptosis. After silencing STAT3 in AML-12 cells, the anti-apoptotic effect of macrophage-derived IL-9 was further enhanced. These results indicate that IL-9 reduces the production of pro-inflammatory factors in ALI. Furthermore, macrophage-derived IL-9 can reduce hepatocyte apoptosis by inhibiting the activation of the STAT3 pathway.


Asunto(s)
Apoptosis , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Etanol/toxicidad , Hepatocitos/patología , Interleucina-9/metabolismo , Macrófagos/inmunología , Factor de Transcripción STAT3/metabolismo , Animales , Depresores del Sistema Nervioso Central/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Interleucina-9/genética , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/genética
19.
Nat Commun ; 12(1): 6773, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799567

RESUMEN

After inoculation by the bite of an infected mosquito, Plasmodium sporozoites enter the blood stream and infect the liver, where each infected cell produces thousands of merozoites. These in turn, infect red blood cells and cause malaria symptoms. To initiate a productive infection, sporozoites must exit the circulation by traversing the blood lining of the liver vessels after which they infect hepatocytes with unique specificity. We screened a phage display library for peptides that structurally mimic (mimotope) a sporozoite ligand for hepatocyte recognition. We identified HP1 (hepatocyte-binding peptide 1) that mimics a ~50 kDa sporozoite ligand (identified as phospholipid scramblase). Further, we show that HP1 interacts with a ~160 kDa hepatocyte membrane putative receptor (identified as carbamoyl-phosphate synthetase 1). Importantly, immunization of mice with the HP1 peptide partially protects them from infection by the rodent parasite P. berghei. Moreover, an antibody to the HP1 mimotope inhibits human parasite P. falciparum infection of human hepatocytes in culture. The sporozoite ligand for hepatocyte invasion is a potential novel pre-erythrocytic vaccine candidate.


Asunto(s)
Vacunas contra la Malaria/uso terapéutico , Malaria Falciparum/prevención & control , Proteínas de Transferencia de Fosfolípidos/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Modelos Animales de Enfermedad , Epítopos/inmunología , Femenino , Células Hep G2 , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/parasitología , Humanos , Hígado/enzimología , Hígado/parasitología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Masculino , Ratones , Biblioteca de Péptidos , Proteínas de Transferencia de Fosfolípidos/aislamiento & purificación , Proteínas de Transferencia de Fosfolípidos/metabolismo , Plasmodium berghei/inmunología , Plasmodium berghei/metabolismo , Plasmodium falciparum/inmunología , Plasmodium falciparum/metabolismo , Cultivo Primario de Células , Proteínas Protozoarias/aislamiento & purificación , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Esporozoítos/metabolismo , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/uso terapéutico
20.
Viruses ; 13(11)2021 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-34835079

RESUMEN

Immunity against hepatitis B virus (HBV) infection is complex and not entirely understood so far, including the decisive factors leading to the development of chronic hepatitis B. This lack of a mechanistic understanding of HBV-specific immunity is also caused by a limited number of suitable animal models. Here, we describe the generation of a recombinant adenovirus expressing an HBV 1.3-overlength genome linked to luciferase (Ad-HBV-Luc) allowing for precise analysis of the quantity of infected hepatocytes. This enables sensitive and close-meshed monitoring of HBV-specific CD8 T cells and the onset of anti-viral immunity in mice. A high dose of Ad-HBV-Luc developed into chronic hepatitis B accompanied by dysfunctional CD8 T cells characterized by high expression of PD1 and TOX and low expression of KLRG1 and GzmB. In contrast, a low dose of Ad-HBV-Luc infection resulted in acute hepatitis with CD8 T cell-mediated elimination of HBV-replicating hepatocytes associated with elevated sALT levels and increased numbers of cytotoxic HBV-specific CD8 T cells. Thus, the infectious dose was a critical factor to induce either acute self-limited or chronic HBV infection in mice. Taken together, the new Ad-HBV-Luc vector will allow for highly sensitive and time-resolved analysis of HBV-specific immune responses during acute and chronic infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/inmunología , Replicación Viral/inmunología , Adenoviridae/genética , Animales , Linfocitos T CD8-positivos/metabolismo , Virus de la Hepatitis B/genética , Hepatitis B Crónica/genética , Hepatitis B Crónica/virología , Hepatocitos/inmunología , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA