Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.497
Filter
1.
J Surg Res ; 296: 18-28, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38215673

ABSTRACT

INTRODUCTION: Ventricular septal defect (VSD) is the most common congenital heart malformation in children. This study aimed to investigate potential pathogenic genes associated with Tibetan familial VSD. METHODS: Whole genomic DNA was extracted from eight Tibetan children with VSD and their healthy parents (a total of 16 individuals). Whole-exome sequencing was performed using the Illumina HiSeq platform. After filtration, detection, and annotation, single nucleotide variations and insertion-deletion markers were examined. Comparative evaluations using the Sorting Intolerant from Tolerant, PolyPhen V2, Mutation Taster, and Combined Annotation Dependent Depletion databases were conducted to predict harmful mutant genes associated with the etiology of Tibetan familial VSD. RESULTS: A total of six missense mutations in genetic disease-causing genes associated with the development of Tibetan familial VSD were identified: activin A receptor type II-like 1 (c.652 C > T: p.R218 W), ATPase cation transporting 13A2 (c.1363 C > T: p.R455 W), endoplasmic reticulum aminopeptidase 1 (c.481 G > A: p.G161 R), MRI1 (c.629 G > A: p.R210Q), tumor necrosis factor receptor-associated protein 1 (c.224 G > A: p.R75H), and FBN2 (c.2260 G > A: p.G754S). The Human Gene Mutation Database confirmed activin A receptor type II-like 1, MRI1, and tumor necrosis factor receptor-associated protein 1 as pathogenic mutations, while FBN2 was classified as a probable pathogenic mutation. CONCLUSIONS: This novel study directly screens genetic variations associated with Tibetan familial VSD using whole-exome sequencing, providing new insights into the pathogenesis of VSD.


Subject(s)
Heart Defects, Congenital , Heart Septal Defects, Ventricular , Child , Humans , Exome Sequencing , Tibet , Heart Septal Defects, Ventricular/genetics , Heart Septal Defects, Ventricular/metabolism , Receptors, Tumor Necrosis Factor/genetics
2.
Front Immunol ; 14: 1225704, 2023.
Article in English | MEDLINE | ID: mdl-37662920

ABSTRACT

The tumor necrosis factor (TNF) receptor superfamily is a structurally and functionally related group of cell surface receptors that play crucial roles in various cellular processes, including apoptosis, cell survival, and immune regulation. This review paper synthesizes key findings from recent studies, highlighting the importance of clustering in TNF receptor superfamily signaling. We discuss the underlying molecular mechanisms of signaling, the functional consequences of receptor clustering, and potential therapeutic implications of targeting surface structures of receptor complexes.


Subject(s)
Receptors, Tumor Necrosis Factor , Signal Transduction , Receptors, Tumor Necrosis Factor/genetics , Cluster Analysis , Cell Membrane , Apoptosis
3.
Eur J Hum Genet ; 31(11): 1337-1341, 2023 11.
Article in English | MEDLINE | ID: mdl-37670079

ABSTRACT

Amelogenesis imperfecta (AI) is a group of rare genetic conditions characterized by quantitative and/or qualitative tooth enamel alterations. AI can manifest as an isolated trait or as part of a syndrome. Recently, five biallelic disease-causing variants in the RELT gene were identified in 7 families with autosomal recessive amelogenesis imperfecta (ARAI). RELT encodes an orphan receptor in the tumor necrosis factor (TNFR) superfamily expressed during tooth development, with unknown function. Here, we report one Brazilian and two French families with ARAI and a distinctive hypomineralized phenotype with hypoplastic enamel, post-eruptive enamel loss, and occlusal attrition. Using Next Generation Sequencing (NGS), four novel RELT variants were identified (c.120+1G>A, p.(?); c.120+1G>T, p.(?); c.193T>C, p.(Cys65Arg) and c.1260_1263dup, p.(Arg422Glyfs*5)). Our findings extend the knowledge of ARAI dental phenotypes and expand the disease-causing variants spectrum of the RELT gene.


Subject(s)
Amelogenesis Imperfecta , Humans , Amelogenesis Imperfecta/genetics , Amelogenesis Imperfecta/pathology , Receptors, Tumor Necrosis Factor/genetics , Phenotype , Brazil , Pedigree
4.
Sci Adv ; 9(23): eadd4977, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37294765

ABSTRACT

It is well established that tumor necrosis factor (TNF) plays an instrumental role in orchestrating the metabolic disorders associated with late stages of cancers. However, it is not clear whether TNF/TNF receptor (TNFR) signaling controls energy homeostasis in healthy individuals. Here, we show that the highly conserved Drosophila TNFR, Wengen (Wgn), is required in the enterocytes (ECs) of the adult gut to restrict lipid catabolism, suppress immune activity, and maintain tissue homeostasis. Wgn limits autophagy-dependent lipolysis by restricting cytoplasmic levels of the TNFR effector, TNFR-associated factor 3 (dTRAF3), while it suppresses immune processes through inhibition of the dTAK1/TAK1-Relish/NF-κB pathway in a dTRAF2-dependent manner. Knocking down dTRAF3 or overexpressing dTRAF2 is sufficient to suppress infection-induced lipid depletion and immune activation, respectively, showing that Wgn/TNFR functions as an intersection between metabolism and immunity allowing pathogen-induced metabolic reprogramming to fuel the energetically costly task of combatting an infection.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , NF-kappa B/metabolism , Energy Metabolism , Lipids , MAP Kinase Kinase Kinases/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
5.
Genes (Basel) ; 14(5)2023 04 28.
Article in English | MEDLINE | ID: mdl-37239365

ABSTRACT

TNF α-induced protein 1 (TNFAIP1) was first identified in human umbilical vein endothelial cells and can be induced by tumor necrosis factor α (TNFα). Early studies have found that TNFAIP1 is involved in the development of many tumors and is closely associated with the neurological disorder Alzheimer's disease. However, little is known about the expression pattern of TNFAIP1 under physiological conditions and its function during embryonic development. In this study, we used zebrafish as a model to illustrate the early developmental expression pattern of tnfaip1 and its role in early development. First, we examined the expression pattern of tnfaip1 during early zebrafish development using quantitative real-time PCR and whole mount in situ hybridization and found that tnfaip1 was highly expressed in early embryonic development and, subsequently, expression became localized to anterior embryonic structures. To investigate the function of tnfaip1 during early development, we constructed a model of a stably inherited tnfaip1 mutant using the CRISPR/Cas9 system. Tnfaip1 mutant embryos showed significant developmental delays as well as microcephaly and microphthalmia. At the same time, we found decreased expression of the neuronal marker genes tuba1b, neurod1, and ccnd1 in tnfaip1 mutants. Analysis of transcriptome sequencing data revealed altered expression of the embryonic development related genes dhx40, hspa13, tnfrsf19, nppa, lrp2b, hspb9, clul1, zbtb47a, cryba1a, and adgrg4a in the tnfaip1 mutants. These findings suggest an important role for tnfaip1 in the early development of zebrafish.


Subject(s)
Adaptor Proteins, Signal Transducing , Neoplasms , Zebrafish Proteins , Zebrafish , Animals , Adaptor Proteins, Signal Transducing/genetics , CRISPR-Cas Systems , Eye Proteins/genetics , Neoplasms/genetics , Receptors, Tumor Necrosis Factor/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Mol Cancer Res ; 21(2): 170-186, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36214671

ABSTRACT

Disease recurrence in high-grade serous ovarian cancer may be due to cancer stem-like cells (CSC) that are resistant to chemotherapy and capable of reestablishing heterogeneous tumors. The alternative NF-κB signaling pathway is implicated in this process; however, the mechanism is unknown. Here we show that TNF-like weak inducer of apoptosis (TWEAK) and its receptor, Fn14, are strong inducers of alternative NF-κB signaling and are enriched in ovarian tumors following chemotherapy treatment. We further show that TWEAK enhances spheroid formation ability, asymmetric division capacity, and expression of SOX2 and epithelial-to-mesenchymal transition genes VIM and ZEB1 in ovarian cancer cells, phenotypes that are enhanced when TWEAK is combined with carboplatin. Moreover, TWEAK in combination with chemotherapy induces expression of the CSC marker CD117 in CD117- cells. Blocking the TWEAK-Fn14-RelB signaling cascade with a small-molecule inhibitor of Fn14 prolongs survival following carboplatin chemotherapy in a mouse model of ovarian cancer. These data provide new insights into ovarian cancer CSC biology and highlight a signaling axis that should be explored for therapeutic development. IMPLICATIONS: This study identifies a unique mechanism for the induction of ovarian cancer stem cells that may serve as a novel therapeutic target for preventing relapse.


Subject(s)
NF-kappa B , Ovarian Neoplasms , Humans , Animals , Female , Mice , NF-kappa B/metabolism , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism , Carboplatin/pharmacology , Receptors, Tumor Necrosis Factor/genetics , TWEAK Receptor/genetics , Cell Line, Tumor , Neoplasm Recurrence, Local/drug therapy , Cytokine TWEAK , Signal Transduction/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Stem Cells/metabolism , Transcription Factor RelB/metabolism
7.
Int J Mol Sci ; 23(19)2022 Oct 02.
Article in English | MEDLINE | ID: mdl-36232986

ABSTRACT

In this study, we presented an AISID method extending AlphaFold-Multimer's success in structure prediction towards identifying specific protein interactions with an optimized AISIDscore. The method was tested to identify the binding proteins in 18 human TNFSF (Tumor Necrosis Factor superfamily) members for each of 27 human TNFRSF (TNF receptor superfamily) members. For each TNFRSF member, we ranked the AISIDscore among the 18 TNFSF members. The correct pairing resulted in the highest AISIDscore for 13 out of 24 TNFRSF members which have known interactions with TNFSF members. Out of the 33 correct pairing between TNFSF and TNFRSF members, 28 pairs could be found in the top five (including 25 pairs in the top three) seats in the AISIDscore ranking. Surprisingly, the specific interactions between TNFSF10 (TNF-related apoptosis-inducing ligand, TRAIL) and its decoy receptors DcR1 and DcR2 gave the highest AISIDscore in the list, while the structures of DcR1 and DcR2 are unknown. The data strongly suggests that AlphaFold-Multimer might be a useful computational screening tool to find novel specific protein bindings. This AISID method may have broad applications in protein biochemistry, extending the application of AlphaFold far beyond structure predictions.


Subject(s)
Receptors, Tumor Necrosis Factor , TNF-Related Apoptosis-Inducing Ligand , Apoptosis , Artificial Intelligence , Humans , Protein Binding , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
BMC Med Genomics ; 15(1): 177, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35941638

ABSTRACT

BACKGROUND: Increasing evidence indicates that the immune microenvironment plays a key role in the genesis and progression of colorectal cancer (CRC). This study aimed to establish an immune-related gene (IRG) signature and determine its clinical prognostic value in patients with CRC. METHODS: The RNA sequencing and associated clinical data of CRC were downloaded from The Cancer Genome Atlas (TCGA) database. We then screened for differentially expressed IRGs by intersecting with IRGs obtained from the Immunology Database and Analysis Portal. Functional enrichment analyses were carried out to determine the potential biological functions and pathways of the IRGs. We also explored the specific molecular mechanisms of the IRGs by constructing regulatory networks. Prognostic IRGs were obtained by LASSO regression analysis, and subsequently, gene models were constructed in the TCGA dataset to confirm the predictive capacity of these IRGs. Finally, we used the TIMER tool to assess the immune properties of prognostic IRGs and correlate them with immune cells. RESULTS: We identified 409 differentially expressed IRGs in patients with CRC. Kyoto Encyclopaedia of Genes and Genomes and Gene Ontology enrichment analyses suggested that these differentially expressed IRGs were significantly related to 102 cancer signalling pathways and various biological functions. Based on the prediction and interaction results, we obtained 59 TF-IRG, 48 miRNA-IRG, and 214 drug-IRG interaction networks for CRC. Four prognostic genes (POMC, TNFRSF19, FGF2, and SCG2) were developed by integrating 47 survival-related IRGs and 42 characteristic CRC genes. The results of gene model showed that patients in the low risk group had better survival outcomes compared to those in the high risk group. The expression of POMC, TNFRSF19, FGF2, and SCG2 was significantly correlated with immune cells. CONCLUSION: This study identified some valid IRGs, and these findings can provide strong evidence for precision immunotherapy in patients with CRC.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Early Detection of Cancer , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Immunotherapy , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Prognosis , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Transcriptome , Tumor Microenvironment
9.
Skelet Muscle ; 12(1): 18, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35902978

ABSTRACT

BACKGROUND: Spinal muscular atrophy (SMA) is a childhood neuromuscular disorder caused by depletion of the survival motor neuron (SMN) protein. SMA is characterized by the selective death of spinal cord motor neurons, leading to progressive muscle wasting. Loss of skeletal muscle in SMA is a combination of denervation-induced muscle atrophy and intrinsic muscle pathologies. Elucidation of the pathways involved is essential to identify the key molecules that contribute to and sustain muscle pathology. The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/TNF receptor superfamily member fibroblast growth factor-inducible 14 (Fn14) pathway has been shown to play a critical role in the regulation of denervation-induced muscle atrophy as well as muscle proliferation, differentiation, and metabolism in adults. However, it is not clear whether this pathway would be important in highly dynamic and developing muscle. METHODS: We thus investigated the potential role of the TWEAK/Fn14 pathway in SMA muscle pathology, using the severe Taiwanese Smn-/-; SMN2 and the less severe Smn2B/- SMA mice, which undergo a progressive neuromuscular decline in the first three post-natal weeks. We also used experimental models of denervation and muscle injury in pre-weaned wild-type (WT) animals and siRNA-mediated knockdown in C2C12 muscle cells to conduct additional mechanistic investigations. RESULTS: Here, we report significantly dysregulated expression of Tweak, Fn14, and previously proposed downstream effectors during disease progression in skeletal muscle of the two SMA mouse models. In addition, siRNA-mediated Smn knockdown in C2C12 myoblasts suggests a genetic interaction between Smn and the TWEAK/Fn14 pathway. Further analyses of SMA, Tweak-/-, and Fn14-/- mice revealed dysregulated myopathy, myogenesis, and glucose metabolism pathways as a common skeletal muscle feature, providing further evidence in support of a relationship between the TWEAK/Fn14 pathway and Smn. Finally, administration of the TWEAK/Fn14 agonist Fc-TWEAK improved disease phenotypes in the two SMA mouse models. CONCLUSIONS: Our study provides mechanistic insights into potential molecular players that contribute to muscle pathology in SMA and into likely differential responses of the TWEAK/Fn14 pathway in developing muscle.


Subject(s)
Muscular Atrophy, Spinal , Receptors, Tumor Necrosis Factor , Animals , Cytokine TWEAK , Disease Models, Animal , Mice , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , RNA, Small Interfering/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Transcription Factors/metabolism
10.
Stem Cell Res Ther ; 13(1): 270, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35729659

ABSTRACT

Stem and progenitor cells (SPCs) possess self-remodeling ability and differentiation potential and are responsible for the regeneration and development of organs and tissue systems. However, the precise mechanisms underlying the regulation of SPC biology remain unclear. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) acts on miscellaneous cells via binding to fibroblast growth factor-inducible 14 (Fn14) and exerts pleiotropic functions in the regulation of divergent stem cell fates. TWEAK/Fn14 signaling can regulate the proliferation, differentiation, and migration of multiple SPCs as well as tumorigenesis in certain contexts. Although TWEAK's roles in modulating multiple SPCs are sparsely reported, the systemic effector functions of this multifaceted protein have not been fully elucidated. In this review, we summarized the fate decisions of TWEAK/Fn14 signaling on multiple stem cells and characterized its potential in stem cell therapy.


Subject(s)
Receptors, Tumor Necrosis Factor , Signal Transduction , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Tumor Necrosis Factor-alpha
11.
Dis Markers ; 2022: 8661423, 2022.
Article in English | MEDLINE | ID: mdl-35707713

ABSTRACT

Background: Lung cancer remains the leading cause of cancer death worldwide, and the most subtype is lung adenocarcinoma (LUAD). Tumor-infiltrating immune cells (TIICs) greatly impact the prognosis of LUAD. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), signal via its receptor fibroblast growth factor-inducible 14 (Fn14), dysregulates immune cell recruitment within tumor environment, thus promoting the progression of autoimmune diseases and cancer. We aimed to explore its role in LUAD. Methods: The expression level of TWEAK was explored in Tumor Immune Estimation Resource 2.0 (TIMER2.0) and Oncomine databases. The Tumor Immune Dysfunction and Exclusion (TIDE) and Lung Cancer Explorer (LCE) databases were applied to evaluate the survival in correlation to TWEAK expression. TIICs were assessed with TIMER2.0 and TIDE datasets. The expression of TWEAK protein was detected in LUAD cell lines and also in tissue samples from LUAD patients via western blotting or combination with immunochemistry. Results: Our results showed that TWEAK was downregulated in LUAD tumors compared to normal tissues in TIMER2.0, Oncomine, cell lines, and clinical specimens. Poor survival was uncovered in lower TWEAK expression of LUAD patients in LCE (meta - HR = 0.84 [95% CI, 0.76-0.92]) and TCGA (Continuous Z = -1.97, p = 0.0486) and GSE13213@PRECOG (Continuous Z = -4.25, p = 2.12e - 5) in TIDE. Multiple tumor-infiltrating immune cells (TIICs) were found closely correlated with TWEAK expression in LUAD, especially hematopoietic stem cell (Rho = 0.505, p = 2.78e - 33), common lymphoid progenitor (Rho = -0.504, p = 3.79e - 33), and myeloid-derived suppressor cells (MDSCs) (Rho = -0.615, p = 1.36e - 52). Conclusion: Lower level of TWEAK was linked with poor survival and aberrant recruitment and phenotype of TIICs in LUAD, which might motivate immune escape and weaken the effects of immunotherapy.


Subject(s)
Adenocarcinoma of Lung , Cytokine TWEAK , Lung Neoplasms , Lymphocytes, Tumor-Infiltrating , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Cytokine TWEAK/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , TWEAK Receptor/genetics , Tumor Necrosis Factors/genetics
12.
J Hepatol ; 77(2): 410-423, 2022 08.
Article in English | MEDLINE | ID: mdl-35351523

ABSTRACT

BACKGROUND & AIMS: The hepatic manifestation of the metabolic syndrome, non-alcoholic fatty liver disease (NAFLD), can lead to the development of hepatocellular carcinoma (HCC). Despite a strong causative link, NAFLD-HCC is often underrepresented in systematic genome explorations. METHODS: Herein, tumor-normal pairs from 100 patients diagnosed with NAFLD-HCC were subject to next-generation sequencing. Bioinformatic analyses were performed to identify key genomic, epigenomic and transcriptomic events associated with the pathogenesis of NAFLD-HCC. Establishment of primary patient-derived NAFLD-HCC culture was used as a representative human model for downstream in vitro investigations of the underlying CTNNB1 S45P driver mutation. A syngeneic immunocompetent mouse model was used to further test the involvement of CTNNB1mutand TNFRSF19 in reshaping the tumor microenvironment. RESULTS: Mutational processes operative in the livers of patients with NAFLD inferred susceptibility to tumor formation through defective DNA repair pathways. Dense promoter mutations and dysregulated transcription factors accentuated activated transcriptional regulation in NAFLD-HCC, in particular the enrichment of MAZ-MYC activities. Somatic events common in HCCs arising from NAFLD and viral hepatitis B infection underscore similar driver pathways, although an incidence shift highlights CTNNB1mut dominance in NAFLD-HCC (33%). Immune exclusion correlated evidently with CTNNB1mut. Chromatin immunoprecipitation-sequencing integrated with transcriptome and immune profiling revealed a unique transcriptional axis, wherein CTNNB1mut leads to an upregulation of TNFRSF19 which subsequently represses senescence-associated secretory phenotype-like cytokines (including IL6 and CXCL8). This phenomenon could be reverted by the Wnt-modulator ICG001. CONCLUSIONS: The unique mutational processes in the livers of patients with NAFLD and NAFLD-HCC allude to a "field effect" involving a gain-of-function role of CTNNB1 mutations in immune exclusion. LAY SUMMARY: The increasing prevalence of metabolic syndrome in adult populations means that NAFLD is poised to be the major cause of liver cancer in the 21st century. We showed a strong "field effect" in the livers of patients with NAFLD, wherein activated ß-catenin was involved in reshaping the tumor-immune microenvironment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Metabolic Syndrome , Non-alcoholic Fatty Liver Disease , Receptors, Tumor Necrosis Factor , beta Catenin , Adult , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Hepatitis B , Humans , Immune Evasion , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mutation , Non-alcoholic Fatty Liver Disease/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Microenvironment , beta Catenin/genetics , beta Catenin/metabolism
13.
Anticancer Res ; 42(3): 1237-1245, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220214

ABSTRACT

BACKGROUND/AIM: Low-grade gliomas (LGG) are heterogenous tumours, causing variable survivals in patients. Identifying molecular markers for a more accurate prognosis is, therefore, important. Since death receptor 6 (DR6) is up-regulated in gliomas and shows an aberrant signalling network, we tested its suitability as a prognostic marker. MATERIALS AND METHODS: DR6 was investigated in patient samples via PCR and western blot. Clinical data were analysed and compared to The Cancer Genome Atlas (TCGA) 'brain lower grade glioma' dataset. RESULTS: DR6 was found to be enhanced in LGG and its expression increased in recurrent LGG. The receptor showed a protective effect in primary LGG with a significantly elongated progression-free survival that was confirmed in the TCGA study. This effect was reversed in relapsed LGG in which cases with high DR6 expression reveal a shorter overall survival. CONCLUSION: DR6 is an interesting candidate for further studies regarding its effect as a prognostic marker, playing an opposing role in primary and relapsed LGG.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Glioma/genetics , Receptors, Tumor Necrosis Factor/genetics , Adult , Biomarkers, Tumor/metabolism , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Databases, Genetic , Female , Glioma/metabolism , Glioma/pathology , Glioma/surgery , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Mutation , Neoplasm Grading , Progression-Free Survival , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
14.
J Med Virol ; 94(2): 480-490, 2022 02.
Article in English | MEDLINE | ID: mdl-31017674

ABSTRACT

Chandipura virus (CHPV) is a neurotropic virus, known to cause encephalitis in humans. The microRNAs (miRNA/miR) play an important role in the pathogenesis of viral infection. The present study is focused on the role of miRNAs during CHPV (strain 1653514) infection in human microglial cells. The deep sequencing of CHPV-infected human microglial cells identified a total of 12 differentially expressed miRNA (DEMs). To elucidate the role of DEMs, the target gene prediction, Gene Ontology term (GO Term), pathway enrichment analysis, and miRNA-messenger RNA (mRNA) interaction network analysis was performed. The GO terms and pathway enrichment analysis provided 146 enriched genes; which were involved in interferon response, cytokine and chemokine signaling. Further, the WGCNA (weighted gene coexpression network analysis) of the enriched genes were discretely categorized into three modules (blue, brown, and turquoise). The hub genes in the blue module may correlate to CHPV induced neuroinflammation. Altogether, the miRNA-mRNA interaction network and WGCNA study revealed the following pairs, hsa-miR-542-3p and FAF1, hsa-miR-92a-1-5p and MYD88, and hsa-miR-3187-3p and TNFRSF21, which may contribute to neuroinflammation during CHPV infection in human microglial cells.


Subject(s)
Gene Regulatory Networks/genetics , MicroRNAs/genetics , Microglia/metabolism , Vesiculovirus/physiology , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Humans , MicroRNAs/metabolism , Myeloid Differentiation Factor 88/genetics , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/virology , Receptors, Tumor Necrosis Factor/genetics , Rhabdoviridae Infections/genetics , Rhabdoviridae Infections/virology
15.
Front Immunol ; 12: 745769, 2021.
Article in English | MEDLINE | ID: mdl-34867972

ABSTRACT

Tumor necrosis factor (TNF) family members participate in the body's antitumor immunity response and influence tumor prognosis and treatment response. However, little is known about the roles of TNF family members in small cell lung cancer (SCLC). Therefore, we conducted the first comprehensive investigation of TNF family members in patients with SCLC, with the goal of using them to predict prognosis and chemotherapy benefit. Abnormal genetic alterations and expression of TNF family members were found to be widespread in SCLC patients. Using LASSO Cox regression analysis, we constructed a TNF family-based signature that separated SCLC patients in the training set (n=77) into high- and low-risk groups with distinct survival and chemotherapy benefit, and the signature was well-validated in the validation set (n=137) by RT-qPCR. Importantly, the signature exhibited superior predictive performance and was identified as a novel independent prognostic factor. Additionally, different immune phenotypes were found between the low-risk and high-risk groups, and high-risk patients had higher CMTM6 expression, suggesting that these patients could benefit from therapeutic methods targeting CMTM6. We constructed the first clinically applicable TNF family-based signature for predicting prognosis and chemotherapy benefit for patients with SCLC. The findings reported here provide a new method for predicting the prognosis of SCLC patients and optimizing clinical management.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Tumor Necrosis Factors/genetics , Aged , Area Under Curve , Chemotherapy, Adjuvant , Datasets as Topic , Female , Gene Expression Profiling , Gene Ontology , Humans , Immunophenotyping , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/physiopathology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Prognosis , Proportional Hazards Models , RNA, Messenger/genetics , ROC Curve , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Retrospective Studies , Risk , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/mortality , Small Cell Lung Carcinoma/physiopathology , Tumor Microenvironment/immunology , Tumor Necrosis Factors/biosynthesis
16.
Mol Syst Biol ; 17(11): e10560, 2021 11.
Article in English | MEDLINE | ID: mdl-34806839

ABSTRACT

T-cell responses to infections and cancers are regulated by co-signalling receptors grouped into the binary categories of co-stimulation or co-inhibition. The co-stimulation TNF receptor superfamily (TNFRSF) members 4-1BB, CD27, GITR and OX40 have similar signalling mechanisms raising the question of whether they have similar impacts on T-cell responses. Here, we screened for the quantitative impact of these TNFRSFs on primary human CD8+ T-cell cytokine production. Although both 4-1BB and CD27 increased production, only 4-1BB was able to prolong the duration over which cytokine was produced, and both had only modest effects on antigen sensitivity. An operational model explained these different phenotypes using shared signalling based on the surface expression of 4-1BB being regulated through signalling feedback. The model predicted and experiments confirmed that CD27 co-stimulation increases 4-1BB expression and subsequent 4-1BB co-stimulation. GITR and OX40 displayed only minor effects on their own but, like 4-1BB, CD27 could enhance GITR expression and subsequent GITR co-stimulation. Thus, different co-stimulation receptors can have different quantitative effects allowing for synergy and fine-tuning of T-cell responses.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation , Humans , Receptors, Tumor Necrosis Factor/genetics
17.
Mol Biol Rep ; 48(11): 7379-7385, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34626314

ABSTRACT

BACKGROUND: Cryptochrome 1 (cry1), the core regulator of the circadian clock, is essential for ontogeny and mammalian reproduction. Unlike in other tissues, the cry1 gene have noncircadian functions in spermatogenesis, which implies the unique role of cry1 gene in the development of testis. The role of cry1 during the puberty has not been described yet. This study aimed to explore the relationship between cry1 expression and spermatogenic cell numbers. METHODS AND RESULTS: We analyzed testicular tissues from Hu sheep aged 0-180 days by hematoxylin and eosin staining, measured cry1 and cell proliferation regulatory factors (bricd5, tnfrsf21, cdk1) expression by quantitative real-time PCR and characterized the transcription factor in the 5' flanking region of cry1 gene. The data revealed that the number of spermatocytes and early spermatocytes increased rapidly from 90 to 120 dpp (day postpartum). Correspondingly, there was a marked variation in the cry1 and cell proliferation related genes (bricd5, tnfrsf21, cdk1) mRNA expression in the testes from the age of 90 days to 180 days (p < 0.05). We also identified some transcription factors (tcfl5) related to cell proliferation. CONCLUSIONS: There is a significant causal relationship between the transcription level of cry1 gene in Hu sheep testes and the number of spermatogenic cells. It is speculated that cry1 gene may regulate the proliferation of spermatogenic cells by regulating the expression of cell proliferation related genes such as bricd5, tnfrsf21 and cdk1.


Subject(s)
CDC2 Protein Kinase/genetics , Cryptochromes/genetics , Receptors, Tumor Necrosis Factor/genetics , Sexual Maturation , Spermatogenesis , Testis/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Gene Expression Regulation, Developmental , Male , Sheep , Spermatocytes/metabolism , Spermatocytes/physiology , Testis/growth & development , Testis/physiology
18.
Int J Mol Sci ; 22(19)2021 Oct 03.
Article in English | MEDLINE | ID: mdl-34639063

ABSTRACT

Autophagy is involved in the degradation of melanosomes and the determination of skin color. TLR4 and tumor necrosis factor (TNF) signaling upregulates NF-kB expression, which is involved in the upregulation of mTOR. The activation of mTOR by UV-B exposure results in decreased autophagy, whereas radiofrequency (RF) irradiation decreases TLR4 and TNF receptor (TNFR) expression. We evaluated whether RF decreased skin pigmentation by restoring autophagy by decreasing the expression of TLR4 or TNFR/NF-κB/mTOR in the UV-B-irradiated animal model. UV-B radiation induced the expressions of TNFR, TLR, and NF-κB in the skin, which were all decreased by RF irradiation. RF irradiation also decreased phosphorylated mTOR expression and upregulated autophagy initiation factors such as FIP200, ULK1, ULK2, ATG13, and ATG101 in the UV-B-irradiated skin. Beclin 1 expression and the expression ratio of LC3-I to LC3-II were increased by UV-B/RF irradiation. Furthermore, melanin-containing autophagosomes increased with RF irradiation. Fontana-Masson staining showed that the amount of melanin deposition in the skin was decreased by RF irradiation. This study showed that RF irradiation decreased skin pigmentation by restoring melanosomal autophagy, and that the possible signal pathways which modulate autophagy could be TLR4, TNFR, NF-κB, and mTOR.


Subject(s)
Autophagy/radiation effects , Melanins/biosynthesis , Melanosomes/metabolism , Radio Waves , Skin Pigmentation/radiation effects , Ultraviolet Rays , Biomarkers , Cells, Cultured , Gene Expression Regulation/radiation effects , Humans , Immunohistochemistry , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Skin Pigmentation/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 4/metabolism
19.
Int J Mol Sci ; 22(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34576184

ABSTRACT

Antibody-cytokine fusion proteins (immunocytokines) are gaining importance for cancer therapy, but those products are often limited by systemic toxicity related to the activity of the cytokine payload in circulation and in secondary lymphoid organs. Tumor necrosis factor (TNF) is used as a pro-inflammatory payload to trigger haemorrhagic necrosis and boost anti-cancer immunity at the tumor site. Here we describe a depotentiated version of TNF (carrying the single point mutation I97A), which displayed reduced binding affinity to its cognate receptor tumor necrosis factor receptor 1 (TNFR-1) and lower biocidal activity. The fusion of the TNF(I97A) mutant to the L19 antibody promoted restoration of anti-tumor activity upon accumulation on the cognate antigen, the alternatively spliced EDB domain of fibronectin. In vivo administration of high doses (375 µg/Kg) of the fusion protein showed a potent anti-tumor effect without apparent toxicity compared with the wild type protein. L19-TNFI97A holds promise for the targeted delivery of TNF activity to neoplastic lesions, helping spare normal tissues.


Subject(s)
Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , Cricetulus , Cytokines/genetics , Cytokines/metabolism , Female , Fibronectins/genetics , Fibronectins/metabolism , Fluorescent Antibody Technique , Immunotherapy , Mice, Inbred BALB C , Mutation , Protein Structure, Secondary , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/genetics
20.
Stem Cell Reports ; 16(9): 2379-2394, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34358453

ABSTRACT

The skin epidermis is a highly compartmentalized tissue consisting of a cornifying epithelium called the interfollicular epidermis (IFE) and associated hair follicles (HFs). Several stem cell populations have been described that mark specific compartments in the skin but none of them is specific to the IFE. Here, we identify Troy as a marker of IFE and HF infundibulum basal layer cells in developing and adult human and mouse epidermis. Genetic lineage-tracing experiments demonstrate that Troy-expressing basal cells contribute to long-term renewal of all layers of the cornifying epithelium. Single-cell transcriptomics and organoid assays of Troy-expressing cells, as well as their progeny, confirmed stem cell identity as well as the ability to generate differentiating daughter cells. In conclusion, we define Troy as a marker of epidermal basal cells that govern interfollicular epidermal renewal and cornification.


Subject(s)
Cell Differentiation/genetics , Epidermal Cells/cytology , Epidermal Cells/metabolism , Hair Follicle/embryology , Hair Follicle/metabolism , Organogenesis/genetics , Receptors, Tumor Necrosis Factor/genetics , Animals , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunohistochemistry , Immunophenotyping , Mice , Organoids , Receptors, Tumor Necrosis Factor/metabolism , Single-Cell Analysis/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...