Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.693
Filtrar
1.
Appl Immunohistochem Mol Morphol ; 32(5): 229-232, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38584487

RESUMO

Gastrointestinal stromal tumors (GISTs) are mesenchymal neoplasms with variable behavior characterized by differentiation toward the interstitial cells of Cajal occurring anywhere in the gastrointestinal stromal tract. Frequently, GISTs have fibrous stroma within tumor cell proliferation areas, which is unlike other types of malignant tumors. If this desmoplasia is active, there is a possibility that some sort of transmitter exists between GIST cells and cells related to fibrosis in the tumor cell proliferation areas. Transforming growth factor (TGF)-ß isoforms, particularly TGF-ß1, are critical for fibrosis pathogenesis. TGF-ß1 regulation of myofibroblasts and fibroblasts during fibrosis is well described. The induced fibroblast activation resulting in myofibroblast differentiation has been reported as an important source of collagen, glycoproteins, proteoglycans, and matrix metallopeptidases in wound healing and fibrosis. However, there are a few reports on the relationship between TGF-ß1 and GISTs. This study aims to clarify TGF-ß1 expression in 30 gastric GISTs using immunohistochemistry and reverse transcription polymerase chain reaction (RT-PCR). For comparison, we also enrolled 30 samples of gastric tubular adenocarcinoma (GTAC). We confirmed TGF-ß1 expression (H-score ≥50 points) in 57% of GIST and 13% of GTAC samples, a significant difference between the 2 tumor types ( P =0.001). We examined the TGF-ß1 mRNA expression of 3 representative GIST samples, each having their respective immunostained areas detected by RT-PCR. Finding TGF-ß1 expression may indicate that this cytokine plays a part in the formation of desmoplasia within GIST cell proliferative areas.


Assuntos
Tumores do Estroma Gastrointestinal , Fator de Crescimento Transformador beta1 , Tumores do Estroma Gastrointestinal/metabolismo , Tumores do Estroma Gastrointestinal/patologia , Tumores do Estroma Gastrointestinal/genética , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/genética , Feminino , Masculino , Pessoa de Meia-Idade , Idoso , Imuno-Histoquímica , Regulação Neoplásica da Expressão Gênica , Adulto , Fibrose , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/patologia , Idoso de 80 Anos ou mais
2.
Int J Oncol ; 64(6)2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38666531

RESUMO

Digestive tract cancer is one of the most common types of cancers globally, with ~4.8 million new cases and 3.4 million cancer­associated deaths in 2018, accounting for 26% of cancer incidence and 35% of cancer­related deaths worldwide. S100 protein family is involved in regulating cancer cell proliferation, angiogenesis, epithelial­mesenchymal transition (EMT), metastasis, metabolism and immune microenvironment homeostasis. The critical role of S100 protein family in digestive tract cancer involves complicated mechanisms, such as cancer stemness remodeling, anaerobic glycolysis regulation, tumor­associated macrophage differentiation and EMT. The present study systematically reviewed published studies on the compositions, function and the underlying molecular mechanisms of the S100 family, as well as guidance for diagnosis, treatment and prognosis of digestive tract cancer. Systematic review of the roles and underlying molecular mechanisms of S100 protein family may provide new insight into exploring potential cancer biomarkers and the optimized therapeutic strategies for digestive tract cancer.


Assuntos
Biomarcadores Tumorais , Transição Epitelial-Mesenquimal , Proteínas S100 , Humanos , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/patologia , Regulação Neoplásica da Expressão Gênica , Neovascularização Patológica/metabolismo , Prognóstico , Proteínas S100/metabolismo , Microambiente Tumoral/imunologia
3.
Int Rev Cell Mol Biol ; 385: 41-99, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38663962

RESUMO

Gastrointestinal carcinomas are a group of cancers associated with the digestive system and its accessory organs. The most prevalent cancers related to the gastrointestinal tract are colorectal, gall bladder, gastric, hepatocellular, and esophageal cancers, respectively. Molecular aberrations in different signaling pathways, such as signal transduction systems or developmental pathways are the chief triggering mechanisms in different cancers Though a massive advancement in diagnostic and therapeutic interventions results in improved survival of patients with gastrointestinal cancer; the lower malignancy stages of these carcinomas are comparatively asymptomatic. Various gastrointestinal-related cancers are detected at advanced stages, leading to deplorable prognoses and increased rates of recurrence. Recent molecular studies have elucidated the imperative roles of several signaling pathways, namely Wnt, Hedgehog, and Notch signaling pathways, play in the progression, therapeutic responsiveness, and metastasis of gastrointestinal-related cancers. This book chapter gives an interesting update on recent findings on the involvement of developmental signaling pathways their mechanistic insight in gastrointestinalcancer. Subsequently, evidences supporting the exploration of gastrointestinal cancer related molecular mechanisms have also been discussed for developing novel therapeutic strategies against these debilitating carcinomas.


Assuntos
Progressão da Doença , Neoplasias Gastrointestinais , Humanos , Neoplasias Gastrointestinais/patologia , Neoplasias Gastrointestinais/metabolismo , Animais , Carcinogênese/patologia , Carcinogênese/metabolismo , Transdução de Sinais
4.
Front Immunol ; 15: 1359914, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646539

RESUMO

Worldwide, gastrointestinal (GI) cancer is recognized as one of the leading malignancies diagnosed in both genders, with mortality largely attributed to metastatic dissemination. It has been identified that in GI cancer, a variety of signaling pathways and key molecules are modified, leading to the emergence of an immunotolerance phenotype. Such modifications are pivotal in the malignancy's evasion of immune detection. Thus, a thorough analysis of the pathways and molecules contributing to GI cancer's immunotolerance is vital for advancing our comprehension and propelling the creation of efficacious pharmacological treatments. In response to this necessity, our review illuminates a selection of groundbreaking cellular signaling pathways associated with immunotolerance in GI cancer, including the Phosphoinositide 3-kinases/Akt, Janus kinase/Signal Transducer and Activator of Transcription 3, Nuclear Factor kappa-light-chain-enhancer of activated B cells, Transforming Growth Factor-beta/Smad, Notch, Programmed Death-1/Programmed Death-Ligand 1, and Wingless and INT-1/beta-catenin-Interleukin 10. Additionally, we examine an array of pertinent molecules like Indoleamine-pyrrole 2,3-dioxygenase, Human Leukocyte Antigen G/E, Glycoprotein A Repetitions Predominant, Clever-1, Interferon regulatory factor 8/Osteopontin, T-cell immunoglobulin and mucin-domain containing-3, Carcinoembryonic antigen-related cell adhesion molecule 1, Cell division control protein 42 homolog, and caspases-1 and -12.


Assuntos
Neoplasias Gastrointestinais , Transdução de Sinais , Humanos , Neoplasias Gastrointestinais/imunologia , Neoplasias Gastrointestinais/patologia , Neoplasias Gastrointestinais/metabolismo , Animais , Metástase Neoplásica , Tolerância Imunológica , Evasão Tumoral
5.
Pediatr Dev Pathol ; 27(3): 228-234, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38512910

RESUMO

INTRODUCTION: Blue rubber bleb nevus syndrome (BRBNS) is an uncommon vascular anomaly characterized by multifocal cutaneous, visceral, and other soft tissue or solid organ venous malformations. We observed that BRBNS lesions express immunohistochemical markers of lymphatic differentiation. METHODS: BRBNS histopathologic specimens assessed at our institution during the past 27 years were reviewed. Slides from 19 BRBNS lesions were selected from 14 patients (9 cutaneous, 9 gastrointestinal, and 1 hepatic). We recorded the involved anatomical compartments and presence/absence of thrombi or vascular smooth muscle. Immunohistochemical endothelial expression of PROX1 (nuclear) and D2-40 (membranous/cytoplasmic) was evaluated semi-quantitatively. RESULTS: Endothelial PROX1 immunopositivity was noted in all specimens; the majority (89.5%) demonstrated staining in more than 10% of cells. D2-40 immunopositivity was present in one-third (33%) of cutaneous lesions and only 1 gastrointestinal lesion. CONCLUSION: Endothelial cells in BRBNS almost always express 1 or more immunohistochemical markers of lymphatic differentiation.


Assuntos
Biomarcadores Tumorais , Neoplasias Gastrointestinais , Imuno-Histoquímica , Nevo Azul , Neoplasias Cutâneas , Humanos , Nevo Azul/metabolismo , Nevo Azul/patologia , Nevo Azul/diagnóstico , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/diagnóstico , Masculino , Criança , Feminino , Pré-Escolar , Adolescente , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/patologia , Neoplasias Gastrointestinais/diagnóstico , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/análise , Lactente , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/análise , Proteínas de Homeodomínio/metabolismo , Endotélio Linfático/metabolismo , Endotélio Linfático/patologia , Anticorpos Monoclonais Murinos/metabolismo
6.
Int J Mol Sci ; 25(5)2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38474232

RESUMO

Gastrointestinal cancers represent one of the more challenging cancers to treat. Current strategies to cure and control gastrointestinal (GI) cancers like surgery, radiation, chemotherapy, and immunotherapy have met with limited success, and research has turned towards further characterizing the tumor microenvironment to develop novel therapeutics. Myeloid-derived suppressor cells (MDSCs) have emerged as crucial drivers of pathogenesis and progression within the tumor microenvironment in GI malignancies. Many MDSCs clinical targets have been defined in preclinical models, that potentially play an integral role in blocking recruitment and expansion, promoting MDSC differentiation into mature myeloid cells, depleting existing MDSCs, altering MDSC metabolic pathways, and directly inhibiting MDSC function. This review article analyzes the role of MDSCs in GI cancers as viable therapeutic targets for gastrointestinal malignancies and reviews the existing clinical trial landscape of recently completed and ongoing clinical studies testing novel therapeutics in GI cancers.


Assuntos
Neoplasias Gastrointestinais , Células Supressoras Mieloides , Humanos , Células Supressoras Mieloides/metabolismo , Neoplasias Gastrointestinais/metabolismo , Células Mieloides , Imunoterapia , Microambiente Tumoral
7.
Commun Biol ; 7(1): 353, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38519773

RESUMO

Cisplatin-based chemotherapy has associated clinical disadvantages, such as high toxicity and resistance. Thus, the development of new antitumor metallodrugs able to overcome different clinical barriers is a public healthcare priority. Here, we studied the mechanism of action of the isomers trans and cis-[PtI2(isopropylamine)2] (I5 and I6, respectively) against gastrointestinal cancer cells. We demonstrate that I5 and I6 modulate mitochondrial metabolism, decreasing OXPHOS activity and negatively affecting ATP-linked oxygen consumption rate. Consequently, I5 and I6 generated Reactive Oxygen Species (ROS), provoking oxidative damage and eventually the induction of senescence. Thus, herein we propose a loop with three interconnected processes modulated by these iodido agents: (i) mitochondrial dysfunction and metabolic disruptions; (ii) ROS generation and oxidative damage; and (iii) cellular senescence. Functionally, I5 reduces cancer cell clonogenicity and tumor growth in a pancreatic xenograft model without systemic toxicity, highlighting a potential anticancer complex that warrants additional pre-clinical studies.


Assuntos
Neoplasias Gastrointestinais , Platina , Humanos , Espécies Reativas de Oxigênio/metabolismo , Cisplatino/farmacologia , Mitocôndrias/metabolismo , Neoplasias Gastrointestinais/metabolismo
8.
Biomed Pharmacother ; 174: 116488, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38520871

RESUMO

Gastrointestinal (GI) cancers have been considered primarily genetic malignancies, caused by a series of progressive genetic alterations. Accumulating evidence shows that histone methylation, an epigenetic modification program, plays an essential role in the different pathological stages of GI cancer progression, such as precancerous lesions, tumorigenesis, and tumor metastasis. Histone methylation-modifying enzymes, including histone methyltransferases (HMTs) and demethylases (HDMs), are the main executor of post-transcriptional modification. The abnormal expression of histone methylation-modifying enzymes characterizes GI cancers with complex pathogenesis and progression. Interactions between upstream controllers and histone methylation-modifying enzymes have recently been revealed, and have provided numerous opportunities to elucidate the pathogenesis of GI cancers in depth and clearly. Here we focus on the association between histone methylation-modifying enzymes and their controllers, aiming to provide a new perspective on the molecular research and clinical management of GI cancers.


Assuntos
Epigênese Genética , Neoplasias Gastrointestinais , Histona Desmetilases , Histonas , Neoplasias Gastrointestinais/patologia , Neoplasias Gastrointestinais/enzimologia , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Humanos , Histonas/metabolismo , Animais , Histona Desmetilases/metabolismo , Histona Metiltransferases/metabolismo , Metilação , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Regulação Neoplásica da Expressão Gênica
9.
J Cancer Res Clin Oncol ; 150(3): 129, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38488909

RESUMO

BACKGROUND: T cells are key players in the tumor immune microenvironment (TIME), as they can recognize and eliminate cancer cells that express neoantigens derived from somatic mutations. However, the diversity and specificity of T-cell receptors (TCRs) that recognize neoantigens are largely unknown, due to the high variability of TCR sequences among individuals. METHODS: To address this challenge, we applied GLIPH2, a novel algorithm that groups TCRs based on their predicted antigen specificity and HLA restriction, to cluster the TCR repertoire of 1,702 patients with digestive tract cancer. The patients were divided into five groups based on whether they carried tumor-infiltrating or clonal-expanded TCRs and calculated their TCR diversity. The prognosis, tumor subtype, gene mutation, gene expression, and immune microenvironment of these groups were compared. Viral specificity inference and immunotherapy relevance analysis performed for the TCR groups. RESULTS: This approach reduced the complexity of TCR sequences to 249 clonally expanded and 150 tumor-infiltrating TCR groups, which revealed distinct patterns of TRBV usage, HLA association, and TCR diversity. In gastric adenocarcinoma (STAD), patients with tumor-infiltrating TCRs (Patients-TI) had significantly worse prognosis than other patients (Patients-nonTI). Patients-TI had richer CD8+ T cells in the immune microenvironment, and their gene expression features were positively correlated with immunotherapy response. We also found that tumor-infiltrating TCR groups were associated with four distinct tumor subtypes, 26 common gene mutations, and 39 gene expression signatures. We discovered that tumor-infiltrating TCRs had cross-reactivity with viral antigens, indicating a possible link between viral infections and tumor immunity. CONCLUSION: By applying GLIPH2 to TCR sequences from digestive tract tumors, we uncovered novel insights into the tumor immune landscape and identified potential candidates for shared TCRs and neoantigens.


Assuntos
Neoplasias Gastrointestinais , Receptores de Antígenos de Linfócitos T , Humanos , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T CD8-Positivos , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Imunoterapia , Antígenos de Neoplasias , Microambiente Tumoral
10.
Hematol Oncol Clin North Am ; 38(3): 585-598, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38521686

RESUMO

The Trastuzumab for Gastric Cancer (ToGA) trial marked a pivotal moment in the adoption of trastuzumab for treating advanced human epidermal growth factor receptor 2 (HER2)-positive esophagogastric (EG) cancer. The KEYNOTE-811 trial brought to light the synergistic effect of immune modulation and HER2 targeting. Additionally, the emergence of trastuzumab deruxtecan (T-DXd) highlighted the potential of new pharmaceutical technologies to extend response, particularly for patients who have advanced beyond initial HER2-targeted therapies. This review aims to navigate through both the successes and challenges encountered historically, as well as promising current trials on innovative and transformative therapeutic strategies, including promising first-in-class and novel first-in-human agents.


Assuntos
Camptotecina/análogos & derivados , Imunoconjugados , Terapia de Alvo Molecular , Receptor ErbB-2 , Humanos , Receptor ErbB-2/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/terapia , Trastuzumab/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Ensaios Clínicos como Assunto
11.
J Proteome Res ; 23(5): 1679-1688, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38546438

RESUMO

Previous metabolomics studies have highlighted the predictive value of metabolites on upper gastrointestinal (UGI) cancer, while most of them ignored the potential effects of lifestyle and genetic risk on plasma metabolites. This study aimed to evaluate the role of lifestyle and genetic risk in the metabolic mechanism of UGI cancer. Differential metabolites of UGI cancer were identified using partial least-squares discriminant analysis and the Wilcoxon test. Then, we calculated the healthy lifestyle index (HLI) score and polygenic risk score (PRS) and divided them into three groups, respectively. A total of 15 metabolites were identified as UGI-cancer-related differential metabolites. The metabolite model (AUC = 0.699) exhibited superior discrimination ability compared to those of the HLI model (AUC = 0.615) and the PRS model (AUC = 0.593). Moreover, subgroup analysis revealed that the metabolite model showed higher discrimination ability for individuals with unhealthy lifestyles compared to that with healthy individuals (AUC = 0.783 vs 0.684). Furthermore, in the genetic risk subgroup analysis, individuals with a genetic predisposition to UGI cancer exhibited the best discriminative performance in the metabolite model (AUC = 0.770). These findings demonstrated the clinical significance of metabolic biomarkers in UGI cancer discrimination, especially in individuals with unhealthy lifestyles and a high genetic risk.


Assuntos
Neoplasias Gastrointestinais , Estilo de Vida Saudável , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/sangue , Reino Unido/epidemiologia , Fatores de Risco , Predisposição Genética para Doença , Bancos de Espécimes Biológicos , Idoso , Metabolômica/métodos , Herança Multifatorial , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/sangue , Estratificação de Risco Genético , Biobanco do Reino Unido
12.
Semin Cancer Biol ; 99: 5-23, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38341121

RESUMO

Gastrointestinal (GI) cancers, including colorectal, gastric, esophageal, pancreatic, and liver, are associated with high mortality and morbidity rates worldwide. One of the underlying reasons for the poor survival outcomes in patients with these malignancies is late disease detection, typically when the tumor has already advanced and potentially spread to distant organs. Increasing evidence indicates that earlier detection of these cancers is associated with improved survival outcomes and, in some cases, allows curative treatments. Consequently, there is a growing interest in the development of molecular biomarkers that offer promise for screening, diagnosis, treatment selection, response assessment, and predicting the prognosis of these cancers. Extracellular vesicles (EVs) are membranous vesicles released from cells containing a repertoire of biological molecules, including nucleic acids, proteins, lipids, and carbohydrates. MicroRNAs (miRNAs) are the most extensively studied non-coding RNAs, and the deregulation of miRNA levels is a feature of cancer cells. EVs miRNAs can serve as messengers for facilitating interactions between tumor cells and the cellular milieu, including immune cells, endothelial cells, and other tumor cells. Furthermore, recent years have witnessed considerable technological advances that have permitted in-depth sequence profiling of these small non-coding RNAs within EVs for their development as promising cancer biomarkers -particularly non-invasive, liquid biopsy markers in various cancers, including GI cancers. Herein, we summarize and discuss the roles of EV-associated miRNAs as they play a seminal role in GI cancer progression, as well as their promising translational and clinical potential as cancer biomarkers as we usher into the area of precision oncology.


Assuntos
Vesículas Extracelulares , Neoplasias Gastrointestinais , MicroRNAs , Humanos , MicroRNAs/genética , Relevância Clínica , Células Endoteliais/metabolismo , Medicina de Precisão , Neoplasias Gastrointestinais/diagnóstico , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Vesículas Extracelulares/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Biologia , Biomarcadores/metabolismo
13.
Br J Cancer ; 130(4): 526-541, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38182686

RESUMO

BACKGROUND: Imatinib has become an exceptionally effective targeted drug for treating gastrointestinal stromal tumors (GISTs). Despite its efficacy, the resistance to imatinib is common in GIST patients, posing a significant challenge to the effective treatment. METHODS: The expression profiling of TRIM21, USP15, and ACSL4 in GIST patients was evaluated using Western blot and immunohistochemistry. To silence gene expression, shRNA was utilized. Biological function of TRIM21, USP15, and ACSL4 was examined through various methods, including resistance index calculation, colony formation, shRNA interference, and xenograft mouse model. The molecular mechanism of TRIM21 and USP15 in GIST was determined by conducting Western blot, co-immunoprecipitation, and quantitative real-time PCR (qPCR) analyses. RESULTS: Here we demonstrated that downregulation of ACSL4 is associated with imatinib (IM) resistance in GIST. Moreover, clinical data showed that higher levels of ACSL4 expression are positively correlated with favorable clinical outcomes. Mechanistic investigations further indicated that the reduced expression of ACSL4 in GIST is attributed to excessive protein degradation mediated by the E3 ligase TRIM21 and the deubiquitinase USP15. CONCLUSION: These findings demonstrate that the TRIM21 and USP15 control ACSL4 stability to maintain the IM sensitive/resistant status of GIST.


Assuntos
Antineoplásicos , Neoplasias Gastrointestinais , Tumores do Estroma Gastrointestinal , Humanos , Animais , Camundongos , Mesilato de Imatinib/farmacologia , Mesilato de Imatinib/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/patologia , Resistencia a Medicamentos Antineoplásicos/genética , RNA Interferente Pequeno/farmacologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Linhagem Celular Tumoral , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Proteases Específicas de Ubiquitina/farmacologia
14.
Front Biosci (Landmark Ed) ; 28(11): 297, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-38062829

RESUMO

Toll-like receptor 3 (TLR3) is a prominent member of the Toll-like receptor (TLR) family and has the ability to recognize and bind intracellular double-stranded RNA (dsRNA). Once triggered by a viral infection or other pathological condition, TLR3 activates immune cells and induces the production of interferons and other immune response molecules. Additionally, TLR3 is considered an important immune modulator, as it can regulate cell apoptosis and promote anticancer immunity. The investigation and application of TLR3 agonists in digestive system tumors have attracted widespread attention and are regarded as a promising cancer treatment strategy with potential clinical applications. TLR3 expression levels are generally elevated in most digestive system tumors, and higher TLR3 expression is associated with a better prognosis. Therefore, TLR3 has emerged as a novel therapeutic target for digestive system tumors. It has been used in combination with chemotherapy, radiotherapy, and targeted therapy and demonstrated excellent efficacy and tolerability. This has provided new ideas and hopes for the treatment of digestive system tumors. This review discusses the mechanisms of TLR3 and its frontier research in digestive system tumors.


Assuntos
Neoplasias do Sistema Digestório , Neoplasias Gastrointestinais , Humanos , Neoplasias do Sistema Digestório/tratamento farmacológico , Neoplasias do Sistema Digestório/genética , Neoplasias do Sistema Digestório/metabolismo , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , RNA de Cadeia Dupla , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/metabolismo , Receptores Toll-Like
15.
Biol Res ; 56(1): 63, 2023 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-38041132

RESUMO

In December 2022 the US Food and Drug Administration (FDA) removed the requirement that drugs in development must undergo animal testing before clinical evaluation, a declaration that now demands the establishment and verification of ex vivo preclinical models that closely represent tumor complexity and that can predict therapeutic response. Fortunately, the emergence of patient-derived organoid (PDOs) culture has enabled the ex vivo mimicking of the pathophysiology of human tumors with the reassembly of tissue-specific features. These features include histopathological variability, molecular expression profiles, genetic and cellular heterogeneity of parental tissue, and furthermore growing evidence suggests the ability to predict patient therapeutic response. Concentrating on the highly lethal and heterogeneous gastrointestinal (GI) tumors, herein we present the state-of-the-art and the current methodology of PDOs. We highlight the potential additions, improvements and testing required to allow the ex vivo of study the tumor microenvironment, as well as offering commentary on the predictive value of clinical response to treatments such as chemotherapy and immunotherapy.


Assuntos
Neoplasias Gastrointestinais , Estados Unidos , Animais , Humanos , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/metabolismo , Organoides/metabolismo , Organoides/patologia , Microambiente Tumoral
16.
Analyst ; 148(22): 5745-5752, 2023 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-37842723

RESUMO

Extracellular vesicles (EVs), as a type of subcellular structure, have been extensively researched for their potential for developing advanced diagnostic technologies for various diseases. However, the biomolecular and biophysical heterogeneity of EVs has restricted their application in clinical settings. In this article, we developed a size-exclusion chromatography-based technique for simultaneous EV size subtyping and protein profiling. By eluting fluorescent aptamer-treated patient plasma through a size-exclusion column, the mixture can be classified into 50 nm aptamer-bound EVs, 100 nm aptamer-bound EVs and free-floating aptamers, which could further enable multiplex EV membrane protein profiling by analyzing the fluorescence intensities of EV-bound aptamers. Using this technique, we successfully identified EV size subtypes for differentiating gastrointestinal cancer prognosis states. Overall, we developed a rapid, user-friendly and low-cost EV size subtyping and protein profiling technique, which holds great potential for identifying crucial EV size subtypes for disease diagnosis in the clinic.


Assuntos
Vesículas Extracelulares , Neoplasias Gastrointestinais , Humanos , Vesículas Extracelulares/química , Cromatografia em Gel , Prognóstico , Neoplasias Gastrointestinais/diagnóstico , Neoplasias Gastrointestinais/metabolismo , Proteínas de Membrana/análise
17.
Crit Rev Oncol Hematol ; 191: 104121, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37690633

RESUMO

Extracellular vesicles (EVs) have gained tremendous interest in the search for next-generation therapeutics for the treatment of a range of pathologies, including cancer, especially due to their small size, biomolecular cargo, ability to mediate intercellular communication, high physicochemical stability, low immunogenicity and biocompatibility. The theranostic potential of EVs have been enhanced by adopting several strategies such as genetic or metabolic engineering, parental cell modification or direct functionalization to incorporate therapeutic compounds into these nanoplatforms. The smart nano-sized EVs indeed offer huge opportunities in the field of cancer, and current research is set at overcoming the existing pitfalls. Smart EVs are already being applied in the clinics despite the challenges faced. We provide, herein, an update on the technologies employed for EV functionalization in order to achieve optimal tumor cell targeting and EV tracking in vivo with bio-imaging modalities, as well as the preclinical and clinical studies making use of these modified EVs, in the context of gastrointestinal tumors.


Assuntos
Vesículas Extracelulares , Neoplasias Gastrointestinais , Humanos , Sistemas de Liberação de Medicamentos/métodos , Medicina de Precisão , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Neoplasias Gastrointestinais/diagnóstico , Neoplasias Gastrointestinais/terapia , Neoplasias Gastrointestinais/metabolismo , Comunicação Celular
18.
Cytokine Growth Factor Rev ; 73: 101-113, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37573251

RESUMO

There is a complex interaction between pro-tumoural and anti-tumoural networks in the tumour microenvironment (TME). Throughout tumourigenesis, communication between malignant cells and various cells of the TME contributes to metabolic reprogramming. Tumour Dysregulation of metabolic pathways offer an evolutional advantage in the TME and enhance the tumour progression, invasiveness, and metastasis. Therefore, understanding these interactions within the TME is crucial for the development of innovative cancer treatments. Extracellular vesicles (EVs) serve as carriers of various materials that include microRNAs, proteins, and lipids that play a vital role in the communication between tumour cells and non-tumour cells. EVs are actively involved in the metabolic reprogramming process. This review summarized recent findings regarding the involvement of EVs in the metabolic reprogramming of various cells in the TME of gastrointestinal cancers. Additionally, we highlight identified microRNAs involved in the reprogramming process in this group of cancers and explained the abnormal tumour metabolism targeted by exosomal cargos as well as the novel potential therapeutic approaches.


Assuntos
Vesículas Extracelulares , Neoplasias Gastrointestinais , MicroRNAs , Neoplasias , Humanos , Comunicação Celular , Neoplasias/metabolismo , Vesículas Extracelulares/fisiologia , MicroRNAs/genética , Neoplasias Gastrointestinais/metabolismo , Carcinogênese/metabolismo , Microambiente Tumoral
19.
Cancer Res ; 83(21): 3624-3635, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37556508

RESUMO

Imatinib mesylate (IM) has revolutionized the treatment of gastrointestinal stromal tumor (GIST). However, most patients inevitably acquire IM resistance. Second- and third-line treatments exhibit modest clinical benefits with a median time to disease progression of 4 to 6 months, highlighting the urgency for novel therapeutic approaches. Here, we report that the expression of BCL6, a known oncogenic driver and transcriptional repressor, was significantly induced in GIST cells following IM treatment. Elevated BCL6 levels suppressed apoptosis and contributed to IM resistance. Mechanistically, BCL6 recruited SIRT1 to the TP53 promoter to modulate histone acetylation and transcriptionally repress TP53 expression. The reduction in p53 subsequently attenuated cell apoptosis and promoted tolerance of GIST cells to IM. Concordantly, treatment of GIST cells showing high BCL6 expression with a BCL6 inhibitor, BI-3802, conferred IM sensitivity. Furthermore, BI-3802 showed striking synergy with IM in IM-responsive and IM-resistant GIST cells in vitro and in vivo. Thus, these findings reveal a role for BCL6 in IM resistance and suggest that a combination of BCL6 inhibitors and IM could be a potentially effective treatment for GIST. SIGNIFICANCE: BCL6 drives resistance to imatinib by inhibiting p53-mediated apoptosis and can be targeted in combination with imatinib to synergistically suppress tumor growth, providing a therapeutic strategy for treating gastrointestinal stromal tumor.


Assuntos
Antineoplásicos , Neoplasias Gastrointestinais , Tumores do Estroma Gastrointestinal , Humanos , Mesilato de Imatinib/farmacologia , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/metabolismo , Proteína Supressora de Tumor p53/genética , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Apoptose , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/genética , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo
20.
Mol Cancer Ther ; 22(8): 926-935, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37141330

RESUMO

Posttranslational modification of proteins increases their diversity and maintains the stability of the intracellular environment. Protein arginine methyltransferases (PRMT) are an important family of epigenetic modification enzymes, which play a critical role in posttranslational modification. In recent years, with the in-depth study of the role of epigenetics, the structure and function of PRMTs have been gradually understood. PRMT enzymatic activity is related to a variety of cellular processes in digestive system malignancies, such as inflammation and immune response, activation of cell cycle and proliferation, inhibition of apoptosis, DNA damage repair, and epithelial-mesenchymal transition. A variety of chemical tools are developed to inhibit PRMT activity, which have been verified by tumor models and clinical trials. This review summarizes the structure and functions of PRMTs as a prelude to our further studies on their role in tumors. The involvement of different PRMTs in the pathogenesis of gastrointestinal tumors is then reviewed. In addition, the application of PRMT inhibitors as therapeutic agents for digestive system cancers is highlighted. In conclusion, PRMTs play an important role in the pathogenesis of gastrointestinal tumors, and their prognostic and therapeutic potential warrants further investigation.


Assuntos
Carcinoma , Neoplasias Gastrointestinais , Humanos , Proteína-Arginina N-Metiltransferases , Neoplasias Gastrointestinais/metabolismo , Carcinoma/metabolismo , Metilação , Sistema Digestório/metabolismo , Arginina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA