Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 661
Filtrar
1.
Science ; 381(6664): 1287-1288, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37733861

RESUMO

Mitochondrial metabolite reduces melanoma growth by boosting antigen presentation.


Assuntos
Apresentação de Antígeno , Melanoma , Mitocôndrias , Humanos , Melanoma/imunologia , Melanoma/patologia , Mitocôndrias/metabolismo , Carcinogênese/imunologia , Carcinogênese/patologia
2.
Front Immunol ; 14: 942849, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36825005

RESUMO

Secretory pathway kinase or kinase-like proteins (SPKKPs) are effective in the lumen of the endoplasmic reticulum (ER), Golgi apparatus (GA), and extracellular space. These proteins are involved in secretory signaling pathways and are distinctive from typical protein kinases. Various reports have shown that SPKKPs regulate the tumorigenesis and progression of human cancer via the phosphorylation of various substrates, which is essential in physiological and pathological processes. Emerging evidence has revealed that the expression of SPKKPs in human cancers is regulated by multiple factors. This review summarizes the current understanding of the contribution of SPKKPs in tumorigenesis and the progression of immunity. With the epidemic trend of immunotherapy, targeting SPKKPs may be a novel approach to anticancer therapy. This study briefly discusses the recent advances regarding SPKKPs.


Assuntos
Neoplasias , Fosfotransferases , Via Secretória , Humanos , Carcinogênese/imunologia , Neoplasias/imunologia , Fosfotransferases/imunologia , Proteínas/imunologia , Via Secretória/imunologia , Transdução de Sinais/imunologia , Progressão da Doença
3.
Nat Commun ; 13(1): 7243, 2022 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-36433954

RESUMO

Exonic circular RNAs (circRNAs) produce predominantly non-coding RNA species that have been recently profiled in many tumors. However, their functional contribution to cancer progression is still poorly understood. Here, we identify the circRNAs expressed in soft tissue sarcoma cells and explore how the circRNAs regulate sarcoma growth in vivo. We show that circCsnk1g3 and circAnkib1 promote tumor growth by shaping a pro-tumorigenic microenvironment, possibly due to their capabilities to regulate tumor-promoting elements extrinsic to the tumor cells. Accordingly, circCsnk1g3 and circAnkib1 can control the expression of interferon-related genes and pro-inflammatory factors in the sarcoma cells, thus directing immune cell recruitment into the tumor mass, and hence their activation. Mechanistically, circRNAs may repress pro-inflammatory elements by buffering activation of the pathways mediated by RIG-I, the cytosolic viral RNA sensor. The current findings suggest that the targeting of specific circRNAs could augment the efficacy of tumor and immune response to mainstay therapies.


Assuntos
Carcinogênese , Interferons , RNA Circular , Sarcoma , Neoplasias de Tecidos Moles , Microambiente Tumoral , Humanos , Carcinogênese/genética , Carcinogênese/imunologia , Interferons/genética , Interferons/imunologia , RNA Circular/genética , RNA Circular/imunologia , Sarcoma/genética , Sarcoma/imunologia , Neoplasias de Tecidos Moles/genética , Neoplasias de Tecidos Moles/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Caseína Quinase I/genética , Caseína Quinase I/imunologia
4.
Proc Natl Acad Sci U S A ; 119(29): e2205574119, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858331

RESUMO

Intestinal barrier immunity is essential for controlling gut microbiota without eliciting harmful immune responses, while its defect contributes to the breakdown of intestinal homeostasis and colitis development. Chemerin, which is abundantly expressed in barrier tissues, has been demonstrated to regulate tissue inflammation via CMKLR1, its functional receptor. Several studies have reported the association between increased expression of chemerin-CMKLR1 and disease severity and immunotherapy resistance in inflammatory bowel disease (IBD) patients. However, the pathophysiological role of endogenous chemerin-CMKLR1 signaling in intestinal homeostasis remains elusive. We herein demonstrated that deficiency of chemerin or intestinal epithelial cell (IEC)-specific CMKLR1 conferred high susceptibility to microbiota-driven neutrophilic colon inflammation and subsequent tumorigenesis in mice following epithelial injury. Unexpectedly, we found that lack of chemerin-CMKLR1 signaling specifically reduced expression of lactoperoxidase (LPO), a peroxidase that is predominantly expressed in colonic ECs and utilizes H2O2 to oxidize thiocyanates to the antibiotic compound, thereby leading to the outgrowth and mucosal invasion of gram-negative bacteria and dysregulated CXCL1/2-mediated neutrophilia. Importantly, decreased LPO expression was causally linked to aggravated microbiota-driven colitis and associated tumorigenesis, as LPO supplementation could completely rescue such phenotypes in mice deficient in epithelial chemerin-CMKLR1 signaling. Moreover, epithelial chemerin-CMKLR1 signaling is necessary for early host defense against bacterial infection in an LPO-dependent manner. Collectively, our study reveals that the chemerin-CMKLR1/LPO axis represents an unrecognized immune mechanism that potentiates epithelial antimicrobial defense and restricts harmful colonic neutrophilia and suggests that LPO supplementation may be beneficial for microbiota dysbiosis in IBD patients with a defective innate antimicrobial mechanism.


Assuntos
Carcinogênese , Quimiocinas , Colite , Colo , Microbioma Gastrointestinal , Peptídeos e Proteínas de Sinalização Intercelular , Lactoperoxidase , Receptores de Quimiocinas , Animais , Carcinogênese/imunologia , Transformação Celular Neoplásica , Quimiocinas/genética , Quimiocinas/metabolismo , Colite/imunologia , Colite/microbiologia , Colo/imunologia , Colo/microbiologia , Peróxido de Hidrogênio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lactoperoxidase/metabolismo , Camundongos , Neutrófilos/imunologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo
5.
Front Immunol ; 13: 891268, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35720407

RESUMO

Since TGF-ß was recognized as an essential secreted cytokine in embryogenesis and adult tissue homeostasis a decade ago, our knowledge of the role of TGF-ß in mammalian development and disease, particularly cancer, has constantly been updated. Mounting evidence has confirmed that TGF-ß is the principal regulator of the immune system, as deprivation of TGF-ß signaling completely abrogates adaptive immunity. However, enhancing TGF-ß signaling constrains the immune response through multiple mechanisms, including boosting Treg cell differentiation and inducing CD8+ T-cell apoptosis in the disease context. The love-hate relationship between TGF-ß signaling and the immune system makes it challenging to develop effective monotherapies targeting TGF-ß, especially for cancer treatment. Nonetheless, recent work on combination therapies of TGF-ß inhibition and immunotherapy have provide insights into the development of TGF-ß-targeted therapies, with favorable outcomes in patients with advanced cancer. Hence, we summarize the entanglement between TGF-ß and the immune system in the developmental and tumor contexts and recent progress on hijacking crucial TGF-ß signaling pathways as an emerging area of cancer therapy.


Assuntos
Neoplasias , Fator de Crescimento Transformador beta , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Carcinogênese/imunologia , Transformação Celular Neoplásica , Humanos , Imunoterapia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Transdução de Sinais/imunologia , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
6.
Nature ; 606(7912): 172-179, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35545680

RESUMO

Missense driver mutations in cancer are concentrated in a few hotspots1. Various mechanisms have been proposed to explain this skew, including biased mutational processes2, phenotypic differences3-6 and immunoediting of neoantigens7,8; however, to our knowledge, no existing model weighs the relative contribution of these features to tumour evolution. We propose a unified theoretical 'free fitness' framework that parsimoniously integrates multimodal genomic, epigenetic, transcriptomic and proteomic data into a biophysical model of the rate-limiting processes underlying the fitness advantage conferred on cancer cells by driver gene mutations. Focusing on TP53, the most mutated gene in cancer1, we present an inference of mutant p53 concentration and demonstrate that TP53 hotspot mutations optimally solve an evolutionary trade-off between oncogenic potential and neoantigen immunogenicity. Our model anticipates patient survival in The Cancer Genome Atlas and patients with lung cancer treated with immunotherapy as well as the age of tumour onset in germline carriers of TP53 variants. The predicted differential immunogenicity between hotspot mutations was validated experimentally in patients with cancer and in a unique large dataset of healthy individuals. Our data indicate that immune selective pressure on TP53 mutations has a smaller role in non-cancerous lesions than in tumours, suggesting that targeted immunotherapy may offer an early prophylactic opportunity for the former. Determining the relative contribution of immunogenicity and oncogenic function to the selective advantage of hotspot mutations thus has important implications for both precision immunotherapies and our understanding of tumour evolution.


Assuntos
Carcinogênese , Evolução Molecular , Neoplasias Pulmonares , Mutação , Carcinogênese/genética , Carcinogênese/imunologia , Conjuntos de Dados como Assunto , Genes p53 , Aptidão Genética , Genômica , Voluntários Saudáveis , Humanos , Imunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Mutação/genética , Mutação de Sentido Incorreto , Reprodutibilidade dos Testes
7.
Mol Biol Rep ; 49(2): 1529-1535, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34981335

RESUMO

Infection processes induce various soluble factors that are carcinogens in humans; therefore, research into the soluble factors of chronic disease released from cells that have been infected with parasites is warranted. Parasitic infections in host cells release high levels of IFNγ. Studies have hypothesised that parasitosis-associated carcinogenesis might be analogous to colorectal cancers developed from inflammatory bowel diseases, whereby various cytokines and chemokines are secreted during chronic inflammation. IL-18 and IL-21 are other factors that might be involved in the development of colorectal cancer in schistosomiasis patients and patients with other infections. IL-21 has profound effects on tumour growth and immunosurveillance of colitis-associated tumourigenesis, thereby emphasising its involvement in the pathogenesis of colorectal cancer. The prominent role of IL-21 in antitumour effects greatly depends on the enhanced cytolytic activity of NK cells and the pathogenic role of IL-21, which is often associated with enhanced risks of cancer and chronic inflammatory processes. As IL-15 is also related to chronic disease, it is believed to also play a role in the antitumour effect of colorectal carcinogenesis. IL-15 generates and maintains long-term CD8+ T cell immunity against T. gondii to control the infection of intracellular pathogens. The lack of IL-15 in mice contributes to the downregulation of the IFNγ-producing CD4+ T cell response against acute T. gondii infection. IL-15 induces hyperplasia and supports the progressive growth of colon cancer via multiple functions. The limited role of IL-15 in the development of NK and CD8+ T cells suggests that there may be other cytokines compensating for the loss of the IL-15 gene.


Assuntos
Neoplasias Colorretais/imunologia , Doenças Transmissíveis/imunologia , Citocinas/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Carcinogênese/imunologia , Carcinogênese/patologia , Colite , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/fisiopatologia , Citocinas/imunologia , Humanos , Inflamação/patologia , Interleucina-15 , Interleucinas , Células Matadoras Naturais/patologia , Camundongos , Toxoplasma , Toxoplasmose/complicações , Toxoplasmose/imunologia
8.
JCI Insight ; 7(2)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35076024

RESUMO

Besides promoting inflammation by mobilizing lipid mediators, group IIA secreted phospholipase A2 (sPLA2-IIA) prevents bacterial infection by degrading bacterial membranes. Here, we show that, despite the restricted intestinal expression of sPLA2-IIA in BALB/c mice, its genetic deletion leads to amelioration of cancer and exacerbation of psoriasis in distal skin. Intestinal expression of sPLA2-IIA is reduced after treatment with antibiotics or under germ-free conditions, suggesting its upregulation by gut microbiota. Metagenome, transcriptome, and metabolome analyses have revealed that sPLA2-IIA deficiency alters the gut microbiota, accompanied by notable changes in the intestinal expression of genes related to immunity and metabolism, as well as in the levels of various blood metabolites and fecal bacterial lipids, suggesting that sPLA2-IIA contributes to shaping of the gut microbiota. The skin phenotypes in Pla2g2a-/- mice are lost (a) when they are cohoused with littermate WT mice, resulting in the mixing of the microbiota between the genotypes, or (b) when they are housed in a more stringent pathogen-free facility, where Pla2g2a expression in WT mice is low and the gut microbial compositions in both genotypes are nearly identical. Thus, our results highlight a potentially new aspect of sPLA2-IIA as a modulator of gut microbiota, perturbation of which affects distal skin responses.


Assuntos
Microbioma Gastrointestinal/imunologia , Fosfolipases A2 do Grupo II/metabolismo , Psoríase , Neoplasias Cutâneas , Animais , Carcinogênese/imunologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Inflamação/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Patologia Molecular/métodos , Psoríase/imunologia , Psoríase/microbiologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/microbiologia
9.
Bioengineered ; 13(2): 3221-3239, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35067176

RESUMO

Secreted phosphoprotein 1 (SPP1) is involved in immune regulation, cell survival, and tumor progression. Studies have demonstrated that SPP1 plays an important role in certain individual tumors. However, the expression profile and oncogenic features of SPP1 in diverse cancers are remaining unknown. Therefore, we performed a comprehensive analysis using The Cancer Genome Atlas (TCGA) database. Raw data of 33 cancer types were download from the University of California Santa Cruz (UCSC) Xena website. The expression of SPP1 and its relationship with tumor prognosis, immune invasion, tumor microenvironment, and immunotherapy were analyzed using the R language. The function analysis was conducted using Gene Set Enrichment Analysis (GSEA). The oncogenic features of SPP1 was validated by wound-healing assay and EdU staining assay. SPP1 highly expressed in most cancers. The expression of SPP1 was significant related to prognosis, tumor mutation burden (TMB), microsatellite instability (MSI), and immune checkpoint genes, suggested that SPP1 plays an essential role in the tumor immune microenvironment and immune cell infiltration. The immune/stromal scores correlated positively with the SPP1 expression, and the relationship was affected by tumor heterogeneity and immunotherapy. In addition, SPP1 could predict the response of tumor immunotherapy. Functional analysis revealed the SPP1-associated terms and pathways. Finally, SPP1 significantly elevated cell proliferation and migration in A549, Huh7, HT-29, A2780 tumor cell lines. In conclusion, this study indicated that SPP1 involved in tumorigenesis, tumor progression, and regulated tumor immune microenvironment, revealing SPP1 might be a potential target for evaluating prognosis and immunotherapy in multiple cancers.


Assuntos
Biomarcadores Tumorais/imunologia , Bases de Dados de Ácidos Nucleicos , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Osteopontina/imunologia , Células A549 , Biomarcadores Tumorais/genética , Carcinogênese/genética , Carcinogênese/imunologia , Feminino , Células HT29 , Humanos , Masculino , Neoplasias/diagnóstico , Neoplasias/genética , Osteopontina/genética
10.
Signal Transduct Target Ther ; 7(1): 19, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35046386

RESUMO

Interleukin-37b (hereafter called IL-37) was identified as fundamental inhibitor of natural and acquired immunity. The molecular mechanism and function of IL-37 in colorectal cancer (CRC) has been elusive. Here, we found that IL-37 transgenic (IL-37tg) mice were highly susceptible to colitis-associated colorectal cancer (CAC) and suffered from dramatically increased tumor burdens in colon. Nevertheless, IL-37 is dispensable for intestinal mutagenesis, and CRC cell proliferation, apoptosis, and migration. Notably, IL-37 dampened protective cytotoxic T cell-mediated immunity in CAC and B16-OVA models. CD8+ T cell dysfunction is defined by reduced retention and activation as well as failure to proliferate and produce cytotoxic cytokines in IL-37tg mice, enabling tumor evasion of immune surveillance. The dysfunction led by IL-37 antagonizes IL-18-induced proliferation and effector function of CD8+ T cells, which was dependent on SIGIRR (single immunoglobulin interleukin-1 receptor-related protein). Finally, we observed that IL-37 levels were significantly increased in CRC patients, and positively correlated with serum CRC biomarker CEA levels, but negatively correlated with the CD8+ T cell infiltration in CRC patients. Our findings highlight the role of IL-37 in harnessing antitumor immunity by inactivation of cytotoxic T cells and establish a new defined inhibitory factor IL-37/SIGIRR in cancer-immunity cycle as therapeutic targets in CRC.


Assuntos
Carcinogênese/imunologia , Colite/imunologia , Neoplasias Colorretais/imunologia , Interleucina-1/imunologia , Proteínas de Neoplasias/imunologia , Receptores de Interleucina-1/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Carcinogênese/genética , Colite/genética , Colite/patologia , Neoplasias Colorretais/genética , Interleucina-1/genética , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Receptores de Interleucina-1/genética
11.
Front Immunol ; 13: 957233, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591314

RESUMO

Introduction: Colorectal cancer and other adult solid cancers pose a significant challenge for successful treatment because the tumor microenvironment both hinders the action of conventional therapeutics and suppresses the immune activities of infiltrating leukocytes. The immune suppression is largely the effect of enhanced local mediators such as purine nucleosides and eicosanoids. Genetic approaches have the promise of interfering with these mechanisms of local immunosuppression to allow both intrinsic and therapeutic immunological anticancer processes. Bacterial phages offer a novel means of enabling access into tissues for therapeutic genetic manipulations. Methods: We generated spheroids of fibroblastic and CRC cancer cells to model the 3-dimensional stromal and parenchymal components of colorectal tumours. We used these to examine the access and effects of both wildtype (WT) and epidermal growth factor (EGF)-presenting bacteriophage λ (WT- λ and EGF-λ) as a means of delivery of targeted genetic interventions in solid cancers. We used both confocal microscopy of spheroids exposed to AF488-tagged phages, and the recovery of viable phages as measured by plaque-forming assays to evaluate access; and measures of mitochondrial enzyme activity and cellular ATP to evaluate the outcome on the constituent cells. Results: Using flourescence-tagged derivatives of these bacteriophages (AF488-WT-λ and AF488-EGF-λ) we showed that phage entry into these tumour microenvironments was possible and that the EGF ligand enabled efficient and persistent uptake into the cancer cell mass. EGF-λ became localized in the intracellular portion of cancer cells and was subjected to subsequent cellular processing. The targeted λ phage had no independent effect upon mature tumour spheroids, but interfered with the early formation and growth of cancer tissues without the need for addition of a toxic payload, suggesting that it might have beneficial effects by itself in addition to any genetic intervention delivered to the tumour. Interference with spheroid formation persisted over the duration of culture. Discussion: We conclude that targeted phage technology is a feasible strategy to facilitate delivery into colorectal cancer tumour tissue (and by extension other solid carcinomas) and provides an appropriate delivery vehicle for a gene therapeutic that can reduce local immunosuppression and/or deliver an additional direct anticancer activity.


Assuntos
Bacteriófago lambda , Carcinogênese , Neoplasias Colorretais , Microambiente Tumoral , Humanos , Bacteriófago lambda/genética , Bacteriófago lambda/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/imunologia , Receptores ErbB/genética , Receptores ErbB/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Carcinogênese/genética , Carcinogênese/imunologia
12.
Nat Immunol ; 23(1): 99-108, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34937926

RESUMO

Enzymes of the TET family are methylcytosine dioxygenases that undergo frequent mutational or functional inactivation in human cancers. Recurrent loss-of-function mutations in TET proteins are frequent in human diffuse large B cell lymphoma (DLBCL). Here, we investigate the role of TET proteins in B cell homeostasis and development of B cell lymphomas with features of DLBCL. We show that deletion of Tet2 and Tet3 genes in mature B cells in mice perturbs B cell homeostasis and results in spontaneous development of germinal center (GC)-derived B cell lymphomas with increased G-quadruplexes and R-loops. At a genome-wide level, G-quadruplexes and R-loops were associated with increased DNA double-strand breaks (DSBs) at immunoglobulin switch regions. Deletion of the DNA methyltransferase DNMT1 in TET-deficient B cells prevented expansion of GC B cells, diminished the accumulation of G-quadruplexes and R-loops and delayed B lymphoma development, consistent with the opposing functions of DNMT and TET enzymes in DNA methylation and demethylation. Clustered regularly interspaced short palindromic repeats (CRISPR)-mediated depletion of nucleases and helicases that regulate G-quadruplexes and R-loops decreased the viability of TET-deficient B cells. Our studies suggest a molecular mechanism by which TET loss of function might predispose to the development of B cell malignancies.


Assuntos
Linfócitos B/imunologia , Carcinogênese/imunologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/imunologia , Dioxigenases/imunologia , Homeostase/imunologia , Estruturas R-Loop/imunologia , Animais , Diferenciação Celular/imunologia , Metilação de DNA/imunologia , Quadruplex G , Centro Germinativo/imunologia , Camundongos , Camundongos Endogâmicos C57BL
13.
FEBS Lett ; 596(4): 427-436, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34939667

RESUMO

T helper cells, especially Th1 and Th17 cells, were reported to play a pivotal role in the pathogenesis of inflammatory bowel disease (IBD). However, the underlying factors regulating T cell functions in IBD progression remain to be fully elucidated. Here, we revealed that IL-17RD/Sef exacerbates DSS-induced colitis by regulating the balance of T cell subsets and their secretion of associated cytokines. We also observed that IL-17RD/Sef promotes colitis-associated tumorigenesis and negatively correlates with survival in both mouse and colorectal cancer patients. Our results suggested that IL-17RD/Sef functions as a regulator of T cell subsets to promote the inflammatory responses in the pathogenesis of IBD and colitis-associated colon cancer.


Assuntos
Carcinogênese/imunologia , Colite/imunologia , Proteínas de Membrana/imunologia , Células Th1/imunologia , Células Th17/imunologia , Animais , Carcinogênese/genética , Carcinogênese/patologia , Colite/induzido quimicamente , Colite/genética , Colite/mortalidade , Colo/imunologia , Colo/patologia , Sulfato de Dextrana/administração & dosagem , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Inflamação , Interferon gama/genética , Interferon gama/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-4/genética , Interleucina-4/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Contagem de Linfócitos , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Análise de Sobrevida , Células Th1/patologia , Células Th17/patologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
14.
Biomed Pharmacother ; 145: 112480, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34915667

RESUMO

Adoptive cell therapy (ACT) based on TCR- or CAR-T cells has become an efficient immunotherapeutic approach for the treatment of various diseases, including cancer. Previously, we developed a novel strategy for generating therapeutic T cell products based on chain-centric TCRs, in which either α- or ß-chain dominates in cognate antigen recognition. To assess the suitability of our experimental approach for the clinical application and predict its possible adverse effects, in studies here, we evaluated the safety of the experimental TCRα-modified T cell product in mouse preclinical models. Our data showed no tumorigenic or mutagenic activity in vitro of TCRα-transduced T cells, indicating no genotoxicity of viral vectors used for the generation of the experimental T cell product. Adoptive transfer of TCRα-engineered T cells in a wide dose range didn`t disturb the host homeostasis and exhibited no acute toxicity or immunotoxicity in vivo. Based on pharmacokinetics and pharmacodynamics analysis here, modified T cells rapidly penetrated and distributed in many viscera after infusion. Histological evaluations revealed no pathological changes in organs caused by T cells accumulation, indicating the absence of non-specific off-target activity or cross-reactivity of the therapeutic TCRα. Studies here provide valuable information on the potential safety of TCRα-T cell based ACT that could be extrapolated to possible effects in a human host.


Assuntos
Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Transferência Adotiva/métodos , Animais , Carcinogênese/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Feminino , Homeostase/imunologia , Humanos , Imunoterapia Adotiva/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese/imunologia
15.
Cancer Lett ; 525: 84-96, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-34740608

RESUMO

Wnt/ß-catenin signaling is a highly conserved pathway that regulates cell proliferation, differentiation, apoptosis, stem cell self-renewal, tissue homeostasis, and wound healing. Dysregulation of the Wnt pathway is intricately involved in almost all stages of tumorigenesis in various cancers. Through direct and/or indirect effects on effector T cells, T-regulatory cells, T-helper cells, dendritic cells, and other cytokine-expressing immune cells, abnormal activation of Wnt/ß-catenin signaling benefits immune exclusion and hinders T-cell-mediated antitumor immune responses. Activation of Wnt signaling results in increased resistance to immunotherapies. In this review, we summarize the process by which Wnt signaling affects cancer and immune surveillance, and the potential for targeting the Wnt-signaling pathway via cancer immunotherapy.


Assuntos
Carcinogênese/genética , Imunoterapia , Neoplasias/imunologia , Microambiente Tumoral/imunologia , Carcinogênese/imunologia , Proliferação de Células/genética , Autorrenovação Celular/imunologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/terapia , Células Th1/imunologia , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/imunologia
16.
Blood ; 139(7): 1052-1065, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34797912

RESUMO

Human T-cell leukemia virus 1 (HTLV-1) causes adult T-cell leukemia (ATL), but the mechanism underlying its initiation remains elusive. In this study, ORP4L was expressed in ATL cells but not in normal T-cells. ORP4L ablation completely blocked T-cell leukemogenesis induced by the HTLV-1 oncoprotein Tax in mice, whereas engineering ORP4L expression in T-cells resulted in T-cell leukemia in mice, suggesting the oncogenic properties and prerequisite of ORP4L promote the initiation of T-cell leukemogenesis. For molecular insight, we found that loss of miR-31 caused by HTLV-1 induced ORP4L expression in T-cells. ORP4L interacts with PI3Kδ to promote PI(3,4,5)P3 generation, contributing to AKT hyperactivation; NF-κB-dependent, p53 inactivation-induced pro-oncogene expression; and T-cell leukemogenesis. Consistently, ORP4L ablation eliminates human ATL cells in patient-derived xenograft ATL models. These results reveal a plausible mechanism of T-cell deterioration by HTLV-1 that can be therapeutically targeted.


Assuntos
Carcinogênese/patologia , Regulação Leucêmica da Expressão Gênica , Infecções por HTLV-I/complicações , Vírus Linfotrópico T Tipo 1 Humano/isolamento & purificação , Leucemia-Linfoma de Células T do Adulto/patologia , Receptores de Esteroides/metabolismo , Linfócitos T/imunologia , Animais , Apoptose , Carcinogênese/imunologia , Carcinogênese/metabolismo , Proliferação de Células , Produtos do Gene tax , Infecções por HTLV-I/virologia , Humanos , Leucemia-Linfoma de Células T do Adulto/genética , Leucemia-Linfoma de Células T do Adulto/metabolismo , Leucemia-Linfoma de Células T do Adulto/virologia , Camundongos , Prognóstico , Receptores de Esteroides/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
FEBS Lett ; 596(4): 449-464, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34855209

RESUMO

The Hippo signaling pathway is critical for carcinogenesis. However, the roles of the Hippo signaling pathway in the tumor immune microenvironment have been rarely investigated. This study systematically analyzed the relationship between the Hippo signaling pathway and immune cell infiltration across 32 cancer types. Both bioinformatics analyses and biological experiments revealed that the downstream effector of Hippo signaling YAP1 might inhibit CD8+ T cell infiltration by upregulating the expression of the transcription factor CREB1 in uterine corpus endometrial carcinoma. In addition, esophageal carcinoma (ESCA) patients were classified into three subtypes based on the Hippo-immune gene panel. The subtypes of ESCA had distinct characteristics in immune cell infiltration, immune pathways, and prognosis. Thus, this study also reveals a new classification of the immune subtypes with prognostic characteristics in ESCA.


Assuntos
Neoplasias do Endométrio/genética , Neoplasias Esofágicas/genética , Via de Sinalização Hippo/genética , Neoplasias/genética , Microambiente Tumoral/genética , Proteínas de Sinalização YAP/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Carcinogênese/genética , Carcinogênese/imunologia , Carcinogênese/patologia , Movimento Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/imunologia , Neoplasias do Endométrio/imunologia , Neoplasias do Endométrio/patologia , Neoplasias Esofágicas/imunologia , Neoplasias Esofágicas/patologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Neoplasias/classificação , Neoplasias/imunologia , Neoplasias/patologia , Prognóstico , Proteômica/métodos , Receptores de Antígenos de Linfócitos B/classificação , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos T/classificação , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Análise de Sobrevida , Terminologia como Assunto , Proteínas de Sinalização YAP/imunologia
18.
Semin Cancer Biol ; 79: 91-104, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34280576

RESUMO

Beyond their well-known functions in the acute phases of the immune response, neutrophils play important roles in the various phases of tumor initiation and progression, through the release of their stored or newly synthesized mediators. In addition to reactive oxygen species, cytokines, chemokines, granule proteins and lipid mediators, neutrophil extracellular traps (NETs) can also be released upon neutrophil activation. NET formation can be achieved through a cell-death process or in association with the release of mitochondrial DNA from viable neutrophils. NETs are described as extracellular fibers of DNA and decorating proteins responsible for trapping and killing extracellular pathogens, playing a protective role in the antimicrobial defense. There is increasing evidence, however, that NETs play multiple roles in the scenario of cancer-related inflammation. For instance, NETs directly or indirectly promote tumor growth and progression, fostering tumor spread at distant sites and shielding cancer cells thus preventing the effects of cytotoxic lymphocytes. NETs can also promote tumor angiogenesis and cancer-associated thrombosis. On the other hand, there is some evidence that NETs may play anti-inflammatory and anti-tumorigenic roles. In this review, we focus on the main mechanisms underlying the emerging effects of NETs in cancer initiation and progression.


Assuntos
Carcinogênese/patologia , Armadilhas Extracelulares/imunologia , Neoplasias/patologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Carcinogênese/imunologia , Citocinas/metabolismo , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Neoplasias/imunologia , Neovascularização Patológica/patologia
19.
Front Immunol ; 12: 793343, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925377

RESUMO

Background: Epigenetics regulate gene expression without altering the DNA sequence. Epigenetics targeted chemotherapeutic approach can be used to overcome treatment resistance and low response rate in HCC. However, a comprehensive review of genomic data was carried out to determine the role of epigenesis in the tumor microenvironment (TME), immune cell-infiltration characteristics in HCC is still insufficient. Methods: The association between epigenetic-related genes (ERGs), inflammatory response-related genes (IRRGs) and CRISPR genes was determined by merging genomic and CRISPR data. Further, characteristics of immune-cell infiltration in the tumor microenvironment was evaluated. Results: Nine differentially expressed genes (ANP32B, ASF1A, BCORL1, BMI1, BUB1, CBX2, CBX3, CDK1, and CDK5) were shown to be independent prognostic factors based on lasso regression in the TCGA-LIHC and ICGC databases. In addition, the results showed significant differences in expression of PDCD-1 (PD-1) and CTLA4 between the high- and low-epigenetic score groups. The CTRP and PRISM-derived drug response data yielded four CTRP-derived compounds (SB-743921, GSK461364, gemcitabine, and paclitaxel) and two PRISM-derived compounds (dolastatin-10 and LY2606368). Patients with high ERGs benefited more from immune checkpoint inhibitor (ICI) therapy than patients with low ERGs. In addition, the high ERGs subgroup had a higher T cell exclusion score, while the low ERGs subgroup had a higher T cell dysfunction. However, there was no difference in microsatellite instability (MSI) score among the two subgroups. Further, genome-wide CRISPR-based loss-of function screening derived from DepMap was conducted to determine key genes leading to HCC development and progression. In total, 640 genes were identified to be essential for survival in HCC cell lines. The protein-protein interaction (PPI) network demonstrated that IRRGs PSEN1 was linked to most ERGs and CRISPR genes such as CDK1, TOP2A, CBX2 and CBX3. Conclusion: Epigenetic alterations of cancer-related genes in the tumor microenvironment play a major role in carcinogenesis. This study showed that epigenetic-related novel biomarkers could be useful in predicting prognosis, clinical diagnosis, and management in HCC.


Assuntos
Carcinoma Hepatocelular/imunologia , Epigênese Genética/genética , Neoplasias Hepáticas/imunologia , Linfócitos T/imunologia , Biomarcadores Tumorais/genética , Carcinogênese/genética , Carcinogênese/imunologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Biologia Computacional , Conjuntos de Dados como Assunto , Epigênese Genética/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Genoma , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Masculino , Proteínas Nucleares/genética , Prognóstico , Análise de Sobrevida , Microambiente Tumoral
20.
J Immunol Res ; 2021: 8512200, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34912899

RESUMO

Large to giant congenital melanocytic nevus (lgCMN) is a benign cutaneous tumor that develops during embryogenesis. A large number of lgCMN patients are ineligible for surgical treatment; hence, there is an urgent need to develop pharmacological treatments. Clinically, tumorigenesis and progression essentially halt after birth, resulting in the homeostasis of growth arrest and survival. Numerous studies have employed whole-genome or whole-exome sequencing to clarify the etiology of lgCMN; however, transcriptome sequencing of lgCMN is still lacking. Through comprehensive transcriptome analysis, this study elucidated the ongoing regulation and homeostasis of lgCMN and identified potential targets for treatment. Transcriptome sequencing, identification of differentially expressed genes and hub genes, protein-protein network construction, functional enrichment, pathway analysis, and gene annotations were performed in this study. Immunohistochemistry, real-time quantitative PCR, immunocytofluorescence, and cell cycle assays were employed for further validation. The results revealed several intriguing phenomena in lgCMN, including P16-induced cell cycle arrest, antiapoptotic activity, and immune evasion caused by malfunction of tumor antigen processing. The arrested cell cycle in lgCMN is consistent with its phenotype and rare malignant transformation. Antiapoptotic activity and immune evasion might explain how such heterogeneous cells have avoided elimination. Major histocompatibility complex (MHC) class I-mediated tumor antigen processing was the hub pathway that was significantly downregulated in lgCMN, and ITCH, FBXW7, HECW2, and WWP1 were identified as candidate hub genes. In conclusion, our research provides new perspectives for immunotherapy and targeted therapy.


Assuntos
Biomarcadores Tumorais/genética , Pontos de Checagem do Ciclo Celular/genética , Nevo Pigmentado/genética , Neoplasias Cutâneas/genética , Evasão Tumoral/genética , Adolescente , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/antagonistas & inibidores , Carcinogênese/genética , Carcinogênese/imunologia , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Imunoterapia/métodos , Lactente , Masculino , Melanócitos , Terapia de Alvo Molecular/métodos , Nevo Pigmentado/imunologia , Nevo Pigmentado/cirurgia , Nevo Pigmentado/terapia , Cultura Primária de Células , Mapas de Interação de Proteínas/genética , Mapas de Interação de Proteínas/imunologia , Pele/patologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/cirurgia , Neoplasias Cutâneas/terapia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...