Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Immunol ; 20(11): 1530-1541, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31591574

RESUMEN

The activation of T cells by the T cell antigen receptor (TCR) results in the formation of signaling protein complexes (signalosomes), the composition of which has not been analyzed at a systems level. Here, we isolated primary CD4+ T cells from 15 gene-targeted mice, each expressing one tagged form of a canonical protein of the TCR-signaling pathway. Using affinity purification coupled with mass spectrometry, we analyzed the composition and dynamics of the signalosomes assembling around each of the tagged proteins over 600 s of TCR engagement. We showed that the TCR signal-transduction network comprises at least 277 unique proteins involved in 366 high-confidence interactions, and that TCR signals diversify extensively at the level of the plasma membrane. Integrating the cellular abundance of the interacting proteins and their interaction stoichiometry provided a quantitative and contextual view of each documented interaction, permitting anticipation of whether ablation of a single interacting protein can impinge on the whole TCR signal-transduction network.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Mapas de Interacción de Proteínas/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Cromatografía de Afinidad/métodos , Espectrometría de Masas/métodos , Ratones , Ratones Transgénicos , Cultivo Primario de Células , Mapeo de Interacción de Proteínas/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/genética
2.
PLoS Pathog ; 18(6): e1010596, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35666747

RESUMEN

Schistosomiasis is caused by parasitic flatworms known as schistosomes and affects over 200 million people worldwide. Prevention of T cell exhaustion by blockade of PD-1 results in clinical benefits to cancer patients and clearance of viral infections, however it remains largely unknown whether loss of PD-1 could prevent or cure schistosomiasis in susceptible mice. In this study, we found that S. japonicum infection dramatically induced PD-1 expression in T cells of the liver where the parasites chronically inhabit and elicit deadly inflammation. Even in mice infected by non-egg-producing unisex parasites, we still observed potent induction of PD-1 in liver T cells of C57BL/6 mice following S. japonicum infection. To determine the function of PD-1 in schistosomiasis, we generated PD-1-deficient mice by CRISPR/Cas9 and found that loss of PD-1 markedly increased T cell count in the liver and spleen of infected mice. IL-4 secreting Th2 cells were significantly decreased in the infected PD-1-deficient mice whereas IFN-γ secreting CD4+ and CD8+ T cells were markedly increased. Surprisingly, such beneficial changes of T cell response did not result in eradication of parasites or in lowering the pathogen burden. In further experiments, we found that loss of PD-1 resulted in both beneficial T cell responses and amplification of regulatory T cells that prevented PD-1-deficient T cells from unleashing anti-parasite activity. Moreover, such PD-1-deficient Tregs exert excessive immunosuppression and express larger amounts of adenosine receptors CD39 and CD73 that are crucial for Treg-mediated immunosuppression. Our experimental results have elucidated the function of PD-1 in schistosomiasis and provide novel insights into prevention and treatment of schistosomiasis on the basis of modulating host adaptive immunity.


Asunto(s)
Schistosoma japonicum , Esquistosomiasis Japónica , Animales , Humanos , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/genética , Linfocitos T Reguladores
3.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33753498

RESUMEN

The homeostasis of protein palmitoylation and depalmitoylation is essential for proper physiological functions in various tissues, in particular the central nervous system (CNS). The dysfunction of PPT1 (PPT1-KI, infantile neuronal ceroid lipofuscinosis [INCL] mouse model), which catalyze the depalmitoylation process, results in serious neurodegeneration accompanied by severe astrogliosis in the brain. Endeavoring to determine critical factors that might account for the pathogenesis in CNS by palm-proteomics, glial fibrillary acidic protein (GFAP) was spotted, indicating that GFAP is probably palmitoylated. Questions concerning if GFAP is indeed palmitoylated in vivo and how palmitoylation of GFAP might participate in neural pathology remain unexplored and are waiting to be investigated. Here we show that GFAP is readily palmitoylated in vitro and in vivo; specifically, cysteine-291 is the unique palmitoylated residue in GFAP. Interestingly, it was found that palmitoylated GFAP promotes astrocyte proliferation in vitro. Furthermore, we showed that PPT1 depalmitoylates GFAP, and the level of palmitoylated GFAP is overwhelmingly up-regulated in PPT1-knockin mice, which lead us to speculate that the elevated level of palmitoylated GFAP might accelerate astrocyte proliferation in vivo and ultimately led to astrogliosis in INCL. Indeed, blocking palmitoylation by mutating cysteine-291 into alanine in GFAP attenuate astrogliosis, and remarkably, the concurrent neurodegenerative pathology in PPT1-knockin mice. Together, these findings demonstrate that hyperpalmitoylated GFAP plays critical roles in regulating the pathogenesis of astrogliosis and neurodegeneration in the CNS, and most importantly, pinpointing that cysteine-291 in GFAP might be a valuable pharmaceutical target for treating INCL and other potential neurodegenerative diseases.


Asunto(s)
Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/metabolismo , Lipofuscinosis Ceroideas Neuronales/metabolismo , Tioléster Hidrolasas/genética , Animales , Astrocitos/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Proteína Ácida Fibrilar de la Glía/genética , Gliosis/genética , Humanos , Lipoilación , Ratones , Ratones Endogámicos C57BL , Lipofuscinosis Ceroideas Neuronales/genética
4.
Immunol Cell Biol ; 100(9): 691-704, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35849045

RESUMEN

Clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) is an efficient tool for establishing genetic models including cellular models, and has facilitated unprecedented advancements in biomedical research. In both patients and cancer animal models, immune cells infiltrate the tumor microenvironment and some of them migrate to draining lymph nodes to exert antitumor effects. Among these immune cells, phagocytes such as macrophages and dendritic cells engulf tumor antigens prior to their crosstalk with T cells and elicit adaptive immune response against tumors. Melanoma cells are frequently used as a tumor model because of their relatively high level of somatic mutations and antigenicity. However, few genetic models have been developed using melanoma cell lines to track tumor cell phagocytosis, which is essential for understanding protective immune response in vivo. In this study, we used CRISPR/Cas9-mediated DNA cleavage and homologous recombination to develop a novel knock-in tool which expresses the ultra-bright fluorescent probe ZsGreen in YUMM1.7 melanoma cells. Using this novel tool, we measured the macrophagic engulfment of melanoma cells inside the tumor microenvironment. We also found that in tumor-grafted mice, a subset of dendritic cells efficiently engulfed YUMM1.7 cells and was preferentially trafficking tumor antigens to draining lymph nodes. In addition, we used this knock-in tool to assess the impact of a point mutation of CD11b on phagocytosis in the tumor microenvironment. Our results demonstrate that the ZsGreen-expressing YUMM1.7 melanoma model provides a valuable tool for the study of phagocytosis in vivo.


Asunto(s)
Antígeno CD11b , Melanoma , Fagocitosis , Animales , Antígenos de Neoplasias , Antígeno CD11b/genética , Línea Celular , Línea Celular Tumoral , Colorantes Fluorescentes , Melanoma/genética , Ratones , Mutación Puntual , Microambiente Tumoral
5.
J Lipid Res ; 60(12): 2006-2019, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31570505

RESUMEN

During foam cell formation and atherosclerosis development, the scavenger receptor CD36 plays critical roles in lipid uptake and triggering of atherogenicity via the activation of Vav molecules. The Vav family includes three highly conserved members known as Vav1, Vav2, and Vav3. As Vav1 and Vav3 were found to exert function in atherosclerosis development, it remains thus to decipher whether Vav2 also plays a role in the development of atherosclerosis. In this study we found that Vav2 deficiency in RAW264.7 macrophages significantly diminished oxidized LDL uptake and CD36 signaling, demonstrating that each Vav protein family member was required for foam cell formation. Genetic disruption of Vav2 in ApoE-deficient C57BL/6 mice significantly inhibited the severity of atherosclerosis. Strikingly, we further found that the genetic deletion of each member of the Vav protein family by CRISPR/Cas9 resulted in a similar alteration of transcriptomic profiles of macrophages. The three members of the Vav proteins were found to form complexes, and genetic ablation of each single Vav molecule was sufficient to prevent endocytosis of CD36. The functional interdependence of the three Vav family members in foam cell formation was due to their indispensable roles in transcriptomic programing, lipid uptake, and activation of the JNK kinase in macrophages.


Asunto(s)
Aterosclerosis/metabolismo , Células Espumosas/citología , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-vav/química , Proteínas Proto-Oncogénicas c-vav/metabolismo , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/genética , Secuencia de Bases , Antígenos CD36/metabolismo , Diferenciación Celular , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos C57BL , Fenotipo , Estructura Cuaternaria de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-vav/deficiencia , Proteínas Proto-Oncogénicas c-vav/genética , Células RAW 264.7
6.
Plant Mol Biol ; 87(1-2): 181-91, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25488398

RESUMEN

As one of the most devastating diseases of rice, sheath blight causes severe rice yield loss. However, little progress has been made in rice breeding for sheath blight resistance. It has been reported that polygalacturonase inhibiting proteins can inhibit the degradation of the plant cell wall by polygalacturonases from pathogens. Here, we prokaryotically expressed and purified OsPGIP1 protein, which was verified by Western blot analysis. Activity assay confirmed the inhibitory activity of OsPGIP1 against the PGase from Rhizoctonia solani. In addition, the location of OsPGIP1 was determined by subcellular localization. Subsequently, we overexpressed OsPGIP1 in Zhonghua 11 (Oryza sativa L. ssp. japonica), and applied PCR and Southern blot analysis to identify the positive T0 transgenic plants with single-copy insertions. Germination assay of the seeds from T1 transgenic plants was carried out to select homozygous OsPGIP1 transgenic lines, and the expression levels of OsPGIP1 in these lines were analyzed by quantitative real-time PCR. Field testing of R. solani inoculation showed that the sheath blight resistance of the transgenic rice was significantly improved. Furthermore, the levels of sheath blight resistance were in accordance with the expression levels of OsPGIP1 in the transgenic lines. Our results reveal the functions of OsPGIP1 and its resistance mechanism to rice sheath blight, which will facilitate rice breeding for sheath blight resistance.


Asunto(s)
Oryza/microbiología , Proteínas de Plantas/fisiología , Rhizoctonia/patogenicidad , Secuencia de Bases , Cartilla de ADN , Genes de Plantas , Oryza/genética , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente , Reacción en Cadena de la Polimerasa
7.
Signal Transduct Target Ther ; 8(1): 236, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37332010

RESUMEN

T lymphopenia, occurring in the early phase of sepsis in response to systemic inflammation, is commonly associated with morbidity and mortality of septic infections. We have previously shown that a sufficient number of T cells is required to constrain Toll-like receptors (TLRs) mediated hyperinflammation. However, the underlying mechanisms remains unsolved. Herein, we unveil that CD4+ T cells engage with MHC II of macrophages to downregulate TLR pro-inflammatory signaling. We show further that the direct contact between CD4 molecule of CD4+ T cells or the ectodomain of CD4 (soluble CD4, sCD4), and MHC II of resident macrophages is necessary and sufficient to prevent TLR4 overactivation in LPS and cecal ligation puncture (CLP) sepsis. sCD4 serum concentrations increase after the onset of LPS sepsis, suggesting its compensatory inhibitive effects on hyperinflammation. sCD4 engagement enables the cytoplasmic domain of MHC II to recruit and activate STING and SHP2, which inhibits IRAK1/Erk and TRAF6/NF-κB activation required for TLR4 inflammation. Furthermore, sCD4 subverts pro-inflammatory plasma membrane anchorage of TLR4 by disruption of MHC II-TLR4 raft domains that promotes MHC II endocytosis. Finally, sCD4/MHCII reversal signaling specifically interferes with TLR4 but not TNFR hyperinflammation, and independent of the inhibitive signaling of CD40 ligand of CD4+ cells on macrophages. Therefore, a sufficient amount of soluble CD4 protein can prevent excessive inflammatory activation of macrophages via alternation of MHC II-TLR signaling complex, that might benefit for a new paradigm of preventive treatment of sepsis.


Asunto(s)
Antígenos CD4 , Sepsis , Humanos , Antígenos CD4/metabolismo , Receptor Toll-Like 4/genética , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Sepsis/genética , Sepsis/metabolismo , Inflamación/metabolismo
8.
Plant Cell Rep ; 31(7): 1173-87, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22362377

RESUMEN

UNLABELLED: Polygalacturonase-inhibiting proteins (PGIPs) are typically leucine-rich repeat (LRR) proteins that can inhibit the activity of fungal polygalacturonases (PGs). In this study, two new Ospgip genes, named Ospgip6 and Ospgip7 with consensus sequence of ten imperfect LRR motif located on rice chromosomes 8 and 9, were identified using BLAST analysis. Both of them appear to be extracellular glycoproteins. To have a global view of the dynamic gene expression pattern, seven Ospgip genes were first analyzed using the Affymetrix rice genome array data from online resource. All of these seven Ospgip genes showed variable expression patterns among tissues/organs. In order to further investigate the potential function of these Ospgip genes, the responses of Ospgip genes to the treatment of various phytohormones (abscisic acid, brassinosteroid, gibberellic acid, 3-indole acetic acid, jasmonic acid, kinetin, naphthalene acetic acid and salicylic acid) as well as fungal infection were analyzed by real-time PCR using time course array. Generally, all the Ospgip genes were slightly up-regulated in the indica rice cultivar Minghui 63 under GA(3), KT and NAA treatments (except Ospgip2, which was down-regulated under KT treatment). In the japonica rice cultivar Zhonghua 11, Ospgip genes were regulated by most treatments with the response time variability. We also analyzed putative cis-elements in the promoter regions of Ospgip genes. This dataset provided a versatile resource to understand the regulatory network of Ospgip genes during the process of phytohormones treatment and fungal infection in the model monocotyledonous plant, rice, and could aid in the transgenic breeding against rice fungal diseases. KEY MESSAGE: All the seven Ospgip genes showed variable expression patterns in Minghui 63 and their expressions were regulated by different phytohormone treatments or fungal infection in Minghui 63 and Zhonghua 11.


Asunto(s)
Oryza/genética , Reguladores del Crecimiento de las Plantas/farmacología , Proteínas de Plantas/metabolismo , Transcriptoma , Secuencia de Aminoácidos , Biología Computacional , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Oryza/metabolismo , Filogenia , Proteínas de Plantas/genética , Regiones Promotoras Genéticas , Rhizoctonia/patogenicidad
9.
Front Cell Infect Microbiol ; 12: 1035765, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36389166

RESUMEN

Schistosomiasis has been widely disseminated around the world, and poses a significant threat to human health. Schistosoma eggs and soluble egg antigen (SEA) mediated inflammatory responses promote the formation of egg granulomas and liver fibrosis. With continuous liver injuries and inflammatory stimulation, liver fibrosis can develop into liver cirrhosis and liver cancer. Therefore, anti-fibrotic therapy is crucial to increase the survival rate of patients. However, current research on antifibrotic treatments for schistosomiasis requires further exploration. In the complicated microenvironment of schistosome infections, it is important to understand the mechanism and pathology of schistosomiasis-associated liver fibrosis(SSLF). In this review, we discuss the role of SEA in inhibiting liver fibrosis, describe its mechanism, and comprehensively explore the role of host-derived and schistosome-derived microRNAs (miRNAs) in SSLF. Inflammasomes and cytokines are significant factors in promoting SSLF, and we discuss the mechanisms of some critical inflammatory signals and pro-fibrotic cytokines. Natural killer(NK) cells and Natural killer T(NKT) cells can inhibit SSLF but are rarely described, therefore, we highlight their significance. This summarizes and provides insights into the mechanisms of key molecules involved in SSLF development.


Asunto(s)
Schistosoma japonicum , Esquistosomiasis Japónica , Esquistosomiasis , Animales , Humanos , Esquistosomiasis Japónica/complicaciones , Esquistosomiasis Japónica/patología , Cirrosis Hepática/patología , Esquistosomiasis/complicaciones , Citocinas
10.
Front Immunol ; 13: 728455, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35769463

RESUMEN

Sphingosine-1-phosphate lyase is encoded by the Sgpl1 gene, degrades S1P, and is crucial for S1P homeostasis in animal models and humans. S1P lyase deficient patients suffer from adrenal insufficiency, severe lymphopenia, and skin disorders. In this study, we used random mutagenesis screening to identify a mouse line carrying a missense mutation of Sgpl1 (M467K). This mutation caused similar pathologies as Sgpl1 knock-out mice in multiple organs, but greatly preserved its lifespan, which M467K mutation mice look normal under SPF conditions for over 40 weeks, in contrast, the knock-out mice live no more than 6 weeks. When treated with Imiquimod, Sgpl1M467K mice experienced exacerbated skin inflammation, as revealed by aggravated acanthosis and orthokeratotic hyperkeratosis. We also demonstrated that the IL17a producing Vγ6+ cell was enriched in Sgpl1M467K skin and caused severe pathology after imiquimod treatment. Interestingly, hyperchromic plaque occurred in the mutant mice one month after Imiquimod treatment but not in the controls, which resembled the skin disorder found in Sgpl1 deficient patients. Therefore, our results demonstrate that Sgpl1M467K point mutation mice successfully modeled a human disease after being treated with Imiquimod. We also revealed a major subset of γδT cells in the skin, IL17 secreting Vγ6 T cells were augmented by Sgpl1 deficiency and led to skin pathology. Therefore, we have, for the first time, linked the IL17a and γδT cells to SPL insufficiency.


Asunto(s)
Hiperpigmentación , Mutación Puntual , Animales , Homeostasis , Imiquimod , Ratones , Ratones Noqueados
11.
Sci China Life Sci ; 64(8): 1227-1235, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33420927

RESUMEN

Neutrophils are crucial for immunity and play important roles in inflammatory diseases; however, mouse models selectively deficient in neutrophils are limited, and neutrophil-specific diphtheria toxin (DT)-based depletion system has not yet been established. In this study, we generated a novel knock-in mouse model expressing diphtheria toxin receptor (DTR) under control of the endogenous Ly6G promoter. We showed that DTR expression was restricted to Ly6G+ neutrophils and complete depletion of neutrophils could be achieved by DT treatment at 24-48 h intervals. We characterized the effects of specific neutrophil depletion in mice at steady-state, with acute inflammation and during tumor growth. Our study presents a valuable new tool to study the roles of neutrophils in the immune system and during tumor progression.


Asunto(s)
Toxina Diftérica/inmunología , Factor de Crecimiento Similar a EGF de Unión a Heparina/inmunología , Neutrófilos/inmunología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología
12.
Atherosclerosis ; 337: 42-52, 2021 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-34757313

RESUMEN

BACKGROUND AND AIMS: Atherosclerosis, a progressive inflammatory disease characterized by elevated inflammation and lipid accumulation in the aortic endothelium, arises in part from the infiltration of inflammatory cells into the vascular wall. However, it is not fully defined how inflammatory cells, especially macrophages, affect the pathogenesis of atherosclerosis. Schlafen4 (Slfn4) mRNA is remarkably upregulated upon ox-LDL stimulation in macrophages. Nonetheless, the role of Slfn4 in foam cell formation remains unclear. METHODS: To determine whether and how Slfn4 regulates lesion macrophage function during atherosclerosis,we engineered ApoE-/-Slfn4-/- double-deficient mice on an ApoE-/- background and evaluated the deficiency of Slfn4 expression in atherosclerotic lesion formation in vivo. RESULTS: Our results demonstrate that total absence of SLFN4 and the bone marrow-restricted deletion of Slfn4 in ApoE-/- mice remarkably diminish inflammatory cell numbers within arterial plaques as well as limit development of atherosclerosis in moderate hypercholesterolemia condition. This is linked to a marked reduction in the expression of proinflammatory cytokines, the generation of the reactive oxygen species (ROS) and the apoptosis of cells. Furthermore, the activation of MAPKs and apoptosis signaling pathways is compromised in the absence of Slfn4. CONCLUSIONS: These findings demonstrate a novel role of Slfn4 in modulating vascular inflammation and atherosclerosis, highlighting a new target for the related diseases.

13.
J Vis Exp ; (177)2021 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-34842230

RESUMEN

Functional genomics studies of the immune system require genetic manipulations that involve both deletion of target genes and addition of elements to proteins of interest. Identification of gene functions in cell line models is important for gene discovery and exploration of cell-intrinsic mechanisms. However, genetic manipulations of immune cells such as T cells and macrophage cell lines using CRISPR/Cas9-mediated knock-in are difficult because of the low transfection efficiency of these cells, especially in a quiescent state. To modify genes in immune cells, drug-resistance selection and viral vectors are typically used to enrich for cells expressing the CRIPSR/Cas9 system, which inevitably results in undesirable intervention of the cells. In a previous study, we designed dual fluorescent reporters coupled to CRISPR/Cas9 that were transiently expressed after electroporation. This technical solution leads to rapid gene deletion in immune cells; however, gene knock-in in immune cells such as T cells and macrophages without the use of drug-resistance selection or viral vectors is even more challenging. In this article, we show that by using cell sorting to aid selection of cells transiently expressing CRISPR/Cas9 constructs targeting the Rosa26 locus in combination with a donor plasmid, gene knock-in can be achieved in both T cells and macrophages without drug-resistance enrichment. As an example, we show how to express human ACE2, a receptor of SARS-Cov-2, which is responsible for the current Covid-19 pandemic, in RAW264.7 macrophages by performing knock-in experiments. Such gene knock-in cells can be widely used for mechanistic studies.


Asunto(s)
COVID-19 , Sistemas CRISPR-Cas , Línea Celular , Técnicas de Sustitución del Gen , Humanos , Macrófagos , Pandemias , SARS-CoV-2 , Linfocitos T
14.
Front Cell Dev Biol ; 9: 769673, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35118065

RESUMEN

Functional genomics in a mammalian model such as mice is fundamental for understanding human biology. The CRISPR/Cas9 system dramatically changed the tempo of obtaining genetic mouse models due to high efficiency. However, experimental evidence for the establishment of sgRNA knock-in animals and analyses of their value in functional genomics are still not sufficient, particularly in mammalian models. In this study, we demonstrate that the establishment of sgRNA knock-in mice is feasible, and more importantly, crosses between sgRNA knock-in mice and the Cas9 constitutively expressing mice result in complete deletion of the target gene. Such sgRNA knock-in provides an alternative approach for in vivo genetic modification and can be useful in multiple circumstances, such as maintenance of genetically modified animals, which are difficult to breed as homozygotes, and cross of such mice to diverse genomic backgrounds to obtain genetically modified animals.

15.
Artículo en Inglés | MEDLINE | ID: mdl-32117051

RESUMEN

Extracellular matrix mineralization is critical for osteogenesis, and its dysregulation could result in osteoporosis and vascular calcification. IKK/NF-κB activation inhibits differentiation of osteoblasts, and reduces extracellular matrix mineralization, however the underlying mechanisms are poorly understood. In this study, we used CRISPR/Cas9 system to permanently inactivate IKKß in preosteoblast cells and confirmed that such cells displayed dramatic increase in extracellular matrix mineralization associated with JNK phosphorylation. Such observation was also found in our study using IKKß-deficient primary murine osteoblasts. Interestingly, we found that in Ikbkb-/-Mapk8-/- or Ikbkb-/-Mapk9-/- double knockout cells, the enhanced mineralization caused by IKKß deficiency was completely abolished, and deletion of either Mapk8 or Mapk9 was sufficient to dampen c-Jun phosphorylation. In further experiments, we discovered that absence of JNK1 or JNK2 on IKKß-deficient background resulted in highly conserved transcriptomic alteration in response to osteogenic induction. Therefore, identification of the indispensable roles of JNK1 and JNK2 in activating c-Jun and promoting osteoblast differentiation on IKKß-deficient background provided novel insights into restoring homeostasis in extracellular matrix mineralization.


Asunto(s)
Calcificación Fisiológica/genética , Matriz Extracelular/metabolismo , Quinasa I-kappa B/genética , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Proteína Quinasa 9 Activada por Mitógenos/fisiología , Osteoblastos/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Células Cultivadas , Matriz Extracelular/genética , Técnicas de Inactivación de Genes , Quinasa I-kappa B/deficiencia , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/genética , Osteoblastos/fisiología , Osteogénesis/genética , Fosforilación/genética , Transducción de Señal/genética
16.
PLoS Negl Trop Dis ; 14(12): e0008909, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33347431

RESUMEN

Schistosomiasis is among the major neglected tropical diseases and effective prevention by boosting the immune system is still not available. T cells are key cellular components governing adaptive immune response to various infections. While common laboratory mice, such as C57BL/6, are highly susceptible to schistosomiasis, the SD rats are extremely resistant. However, whether adaptive immunity is necessary for such natural resistance to schistosomiasis in rats remains to be determined. Therefore, it is necessary to establish genetic model deficient in T cells and adaptive immunity on the resistant SD background, and to characterize liver pathology during schistosomiasis. In this study we compared experimental schistosomiasis in highly susceptible C57BL/6 (B6) mice and in resistant SD rats, using cercariae of Schistosoma japonicum. We observed a marked T cell expansion in the spleen of infected B6 mice, but not resistant SD rats. Interestingly, CD3e-/- B6 mice in which T cells are completely absent, the infectious burden of adult worms was significantly higher than that in WT mice, suggesting an anti-parasitic role for T cells in B6 mice during schistosome infection. In further experiments, we established Lck deficient SD rats by using CRISPR/Cas9 in which T cell development was completely abolished. Strikingly, we found that such Lck deficiency in SD rats severely impaired their natural resistance to schistosome infection, and fostered parasite growth. Together with an additional genetic model deficient in T cells, the CD3e-/- SD rats, we confirmed the absence of T cell resulted in loss of natural resistance to schistosome infection, but also mitigated liver immunopathology. Our further experiments showed that regulatory T cell differentiation in infected SD rats was significantly decreased during schistosomiasis, in contrast to significant increase of regulatory T cells in infected B6 mice. These data suggest that T cell mediated immune tolerance facilitates persistent infection in mice but not in SD rats. The demonstration of an important role for T cells in natural resistance of SD rats to schistosomiasis provides experimental evidences supporting the rationale to boost T cell responses in humans to prevent and treat schistosomiasis.


Asunto(s)
Esquistosomiasis Japónica/inmunología , Linfocitos T/fisiología , Animales , Complejo CD3/genética , Complejo CD3/metabolismo , Sistemas CRISPR-Cas , Eliminación de Gen , Regulación de la Expresión Génica , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Schistosoma japonicum/fisiología
17.
Artículo en Inglés | MEDLINE | ID: mdl-33222673

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is a major subtype of esophageal cancers. The Five-year survival rate of ESCC is low and molecular targets for ESCC treatment and prognosis assessment are very limited. T cells are critical for clearance of cancer cells and blockade of co-inhibitory molecules for T cell activation has emerged as a promising therapy to treat cancer patients. However, in ESCC patients such co-inhibitory molecules regulating T cell activation is poorly documented. OBJECTIVE: We aim to evaluate how the presence of inhibitory check-point molecules in T cells could impact survival of patients. METHODS: We performed follow-up study of 161 patients undergoing resection of esophageal carcinoma from February 2014 to December 2015, by immunohistochemical staining of six co-inhibitory molecules for T cell activation, namely PD-1, CTLA-4, TIM-3, LAG-3, BTLA and A2AR. Expression of each of the six co-inhibitory molecules was analyzed for its correlation with patient survival by Kaplan-Meier survival analysis. We also applied Kaplan-Meier analyses to evaluate concomitant expression of co-inhibitory molecules and their correlation with patient survival. RESULTS: We found that levels of PD-1, TIM-3 and BTLA can be used as independent prognostic factors for overall survival of patients with ESCC. More importantly, our study found that the co-expression of PD-1 and TIM-3, PD-1 and BTLA, TIM-3 and BTLA significantly reduced the survival of patients with ESCC (P<0.05). CONCLUSION: Therefore, our results suggest the necessity of evaluating the tumor tissue expression of co-inhibitory molecules and targeting co-expressed molecules in immunotherapies for ESCC patients.

18.
Mol Immunol ; 128: 219-226, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33157351

RESUMEN

NF-κB activation is essential in mediating the induction of pro-inflammatory cytokines and also plays a key role in regulating the inflammatory response through intricate mechanisms. In this study, loss of Gfi1 was found to be associated with transcriptomic profiles related to NF-κB activation, including an increase in pro-inflammatory cytokines. Genetically inactivating the IKK/NF-κB signaling pathway in macrophages showed that Gfi1 deficiency led to pro-inflammatory cytokine production requiring NF-κB activation. More importantly, we revealed that one of the under-researched mechanisms, involving Gfi1 and Zc3h12c exerted negative regulation on NF-κB activation. Both Gfi1 and Zc3h12c were found to inhibit NF-κB activation, and double knockout exhibited additive roles of Gfi1 and Zc3h12c in preventing proinflammatory cytokine production. The loss of Gfi1 upregulated Zc3h12c which in turn inhibited NF-κB activation. Therefore, this study delineates the function of Zc3h12c in enhancing the negative regulation of Gfi1 through NF-κB activation during inflammation in macrophages.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Retroalimentación , Regulación de la Expresión Génica/fisiología , Ratones , Células RAW 264.7 , Transducción de Señal/fisiología , Activación Transcripcional/fisiología
19.
Front Immunol ; 11: 607442, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488612

RESUMEN

Zdhhc family genes are composed of 24 members that regulate palmitoylation, a post-translational modification process for proteins. Mutations in genes that alter palmitoylation or de-palmitoylation could result in neurodegenerative diseases and inflammatory disorders. In this study, we found that Zdhhc2 was robustly induced in psoriatic skin and loss of Zdhhc2 in mice by CRISPR/Cas9 dramatically inhibited pathology of the ear skin following imiquimod treatment. As psoriasis is an inflammatory disorder, we analyzed tissue infiltrating immune cells and cytokine production. Strikingly we found that a master psoriatic cytokine interferon-α (IFN-α) in the lesioned skin of wildtype (WT) mice was 23-fold higher than that in Zdhhc2 deficient counterparts. In addition, we found that CD45+ white blood cells (WBC) infiltrating in the skin of Zdhhc2 deficient mice were also significantly reduced. Amelioration in psoriasis and dramatically reduced inflammation of Zdhhc2 deficient mice led us to analyze the cellular components that were affected by loss of Zdhhc2. We found that imiquimod induced plasmacytoid dendritic cell (pDC) accumulation in psoriatic skin, spleen, and draining lymph nodes (DLN) were drastically decreased in Zdhhc2 deficient mice, and the expression of pDC activation marker CD80 also exhibited significantly inhibited in psoriatic skin. In further experiments, we confirmed the cell intrinsic effect of Zdhhc2 on pDCs as we found that loss of zDHHC2 in human CAL-1 pDC dampened both interferon regulatory factor 7 (IRF7) phosphorylation and IFN-α production. Therefore, we identified novel function of Zdhhc2 in controlling inflammatory response in psoriasis in mice and we also confirmed that crucial role of Zdhhc2 in pDCs by regulating IRF7 activity and production of the critical cytokine. Our results finding the dependence of IFN-α production on Zdhhc2 in inflamed murine skin and in human pDCs provide rationale for targeting this new molecule in treatment of inflammation.


Asunto(s)
Aciltransferasas/metabolismo , Células Dendríticas/enzimología , Interferón-alfa/metabolismo , Psoriasis/enzimología , Piel/enzimología , Proteínas Supresoras de Tumor/metabolismo , Aciltransferasas/genética , Animales , Línea Celular , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Imiquimod , Interferón-alfa/genética , Activación de Linfocitos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Psoriasis/inducido químicamente , Psoriasis/genética , Psoriasis/inmunología , Transducción de Señal , Piel/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA