Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.076
Filtrer
1.
Cytopathology ; 35(4): 488-496, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38752464

RÉSUMÉ

BACKGROUND: Metastatic lesions to the salivary gland are rare and mostly affect the parotids. Metastases represent 8% of all malignant lesions of the parotid gland. Around 80% originate from squamous cell carcinomas (SCC) of the head and neck region. Fine needle aspiration (FNA) plays a crucial role in distinguishing primary salivary gland lesions from metastases. Herein we describe our series of metastases to the parotid glands. MATERIALS AND METHODS: We analysed 630 parotid gland FNAs over a decade including conventional and liquid-based cytology specimens. Ancillary techniques such as immunocytochemistry (ICC) were conducted on cell blocks. RESULTS: Eighty (12.4%) cases were malignant lesions, of which 53 (63.75%) were metastases including 24% melanoma, 22.6% SCC, 19% renal carcinomas, 7.5% breast carcinomas, 11.3% lung, 9% intestinal and 1.8% testicular, malignant solitary fibrous tumour and Merkel cell carcinoma. The 53 cases, classified according to the Milan system for salivary cytopathology, belonged to 5 Suspicious for malignancy (SFM) and 48 malignant (M) categories. Forty had a known history of primary malignancy (75.4%), while 13 were suspicious to be a metastatic localisation (24.5%), distributed as 5SFM (2SCC and 3Melanoma) and 8 M. A combination of clinical history, cytomorphology and ICC identified 100% of them. CONCLUSIONS: Fine needle aspiration plays a central role in the diagnostic workup of patients with metastatic lesions to their parotid glands, thereby defining the correct management. Diagnostic accuracy may be enhanced by applying ICC. Although melanoma and SCC are the most common histological types, several other malignancies may also metastasize to the parotid glands and should be kept into consideration.


Sujet(s)
Glande parotide , Tumeurs de la parotide , Humains , Femelle , Mâle , Tumeurs de la parotide/anatomopathologie , Tumeurs de la parotide/diagnostic , Tumeurs de la parotide/secondaire , Adulte d'âge moyen , Sujet âgé , Cytoponction/méthodes , Glande parotide/anatomopathologie , Adulte , Sujet âgé de 80 ans ou plus , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/diagnostic , Carcinome épidermoïde/secondaire , Mélanome/anatomopathologie , Mélanome/diagnostic , Métastase tumorale/anatomopathologie , Cytodiagnostic/méthodes , Adolescent
2.
Immunity ; 57(4): 840-842, 2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38599176

RÉSUMÉ

Stress hormones can contribute to cancer progression, but how immune cells play a role in this process is unclear. In a recent study in Cancer Cell, He et al. showed that glucocorticoids potentiate metastasis by skewing neutrophils toward pro-tumorigenic functions.


Sujet(s)
Tumeurs , Granulocytes neutrophiles , Humains , Tumeurs/anatomopathologie , Microenvironnement tumoral , Métastase tumorale/anatomopathologie
3.
Bull Math Biol ; 86(6): 64, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38664343

RÉSUMÉ

We introduce in this paper substantial enhancements to a previously proposed hybrid multiscale cancer invasion modelling framework to better reflect the biological reality and dynamics of cancer. These model updates contribute to a more accurate representation of cancer dynamics, they provide deeper insights and enhance our predictive capabilities. Key updates include the integration of porous medium-like diffusion for the evolution of Epithelial-like Cancer Cells and other essential cellular constituents of the system, more realistic modelling of Epithelial-Mesenchymal Transition and Mesenchymal-Epithelial Transition models with the inclusion of Transforming Growth Factor beta within the tumour microenvironment, and the introduction of Compound Poisson Process in the Stochastic Differential Equations that describe the migration behaviour of the Mesenchymal-like Cancer Cells. Another innovative feature of the model is its extension into a multi-organ metastatic framework. This framework connects various organs through a circulatory network, enabling the study of how cancer cells spread to secondary sites.


Sujet(s)
Transition épithélio-mésenchymateuse , Concepts mathématiques , Modèles biologiques , Invasion tumorale , Métastase tumorale , Tumeurs , Microenvironnement tumoral , Humains , Métastase tumorale/anatomopathologie , Microenvironnement tumoral/physiologie , Transition épithélio-mésenchymateuse/physiologie , Tumeurs/anatomopathologie , Processus stochastiques , Mouvement cellulaire , Facteur de croissance transformant bêta/métabolisme , Simulation numérique , Loi de Poisson
4.
Biomolecules ; 14(2)2024 Feb 03.
Article de Anglais | MEDLINE | ID: mdl-38397421

RÉSUMÉ

Tumor diseases become a huge problem when they embark on a path that advances to malignancy, such as the process of metastasis. Cancer metastasis has been thoroughly investigated from a biological perspective in the past, whereas it has still been less explored from a physical perspective. Until now, the intraluminal pathway of cancer metastasis has received the most attention, while the interaction of cancer cells with macrophages has received little attention. Apart from the biochemical characteristics, tumor treatments also rely on the tumor microenvironment, which is recognized to be immunosuppressive and, as has recently been found, mechanically stimulates cancer cells and thus alters their functions. The review article highlights the interaction of cancer cells with other cells in the vascular metastatic route and discusses the impact of this intercellular interplay on the mechanical characteristics and subsequently on the functionality of cancer cells. For instance, macrophages can guide cancer cells on their intravascular route of cancer metastasis, whereby they can help to circumvent the adverse conditions within blood or lymphatic vessels. Macrophages induce microchannel tunneling that can possibly avoid mechanical forces during extra- and intravasation and reduce the forces within the vascular lumen due to vascular flow. The review article highlights the vascular route of cancer metastasis and discusses the key players in this traditional route. Moreover, the effects of flows during the process of metastasis are presented, and the effects of the microenvironment, such as mechanical influences, are characterized. Finally, the increased knowledge of cancer metastasis opens up new perspectives for cancer treatment.


Sujet(s)
Tumeurs , Microenvironnement tumoral , Humains , Signaux , Tumeurs/métabolisme , Macrophages/métabolisme , Phénomènes mécaniques , Métastase tumorale/anatomopathologie
5.
J Vis Exp ; (203)2024 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-38345216

RÉSUMÉ

Ovarian cancer is the deadliest gynecologic malignancy. The omentum plays a key role in providing a supportive microenvironment to metastatic ovarian cancer cells as well as immune modulatory signals that allow tumor tolerance. However, we have limited models that closely mimic the interaction between ovarian cancer cells and adipose-rich tissues. To further understand the cellular and molecular mechanisms by which the omentum provides a pro-tumoral microenvironment, we developed a unique 3D ex vivo model of cancer cell-omentum interaction. Using human omentum, we are able to grow ovarian cancer cells within this adipose-rich microenvironment and monitor the factors responsible for tumor growth and immune regulation. In addition to providing a platform for the study of this adipose-rich tumor microenvironment, the model provides an excellent platform for the development and evaluation of novel therapeutic approaches to target metastatic cancer cells in this niche. The proposed model is easy to generate, inexpensive, and applicable to translational investigations.


Sujet(s)
Tumeurs de l'ovaire , Tumeurs du péritoine , Humains , Femelle , Tumeurs du péritoine/secondaire , Omentum , Tumeurs de l'ovaire/anatomopathologie , Tissu adipeux/anatomopathologie , Métastase tumorale/anatomopathologie , Microenvironnement tumoral
6.
Platelets ; 35(1): 2315037, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38372252

RÉSUMÉ

Glycosylation is a ubiquitous cellular or microenvironment-specific post-translational modification that occurs on the surface of normal cells and tumor cells. Tumor cell-associated glycosylation is involved in hematogenous metastasis. A wide variety of tumors undergo aberrant glycosylation to interact with platelets. As platelets have many opportunities to engage circulating tumor cells, they represent an important avenue into understanding the role glycosylation plays in tumor metastasis. Platelet involvement in tumor metastasis is evidenced by observations that platelets protect tumor cells from damaging shear forces and immune system attack, aid metastasis through the endothelium at specific sites, and facilitate tumor survival and colonization. During platelet-tumor-cell interactions, many opportunities for glycan-ligand binding emerge. This review integrates the latest information about glycans, their ligands, and how they mediate platelet-tumor interactions. We also discuss adaptive changes that tumors undergo upon glycan-lectin binding and the impact glycans have on targeted therapeutic strategies for treating tumors in clinical settings.


Tumor hematogenous metastasis is a serious threat to the survival and prognosis of patients, and a variety of factors help this process to occur, and platelets are also involved. During tumor cell metastasis, platelets can adhere to each other and tumor cells, a phenomenon that leads to the immunity of tumor cells from various threats in metastasis, including immune attacks, shearing forces, etc. Scientists have shown that the adhesion effect between platelets and tumor cells is often dependent on various types of sugars, which are not the sugars we ingest. These sugars often appear as glycosylation modifications on the proteins of the cells, including normal glycosylation modifications and some abnormal structures that only appear on tumor cells, and their ligands, lectins, are also present on the surface of the tumor cells or platelets. Their combination results in the better adaptation of tumor cells to the metastatic process, where proteins such as P-selectin, CLEC-2, and Galectins have been more studied. Focusing on Glycan-Lectin interactions between platelets and tumor cells, related studies help us to further understand tumor metastasis, and intervene in this binding and develop related drugs with great potential.


Sujet(s)
Lectines , Tumeurs , Humains , Lectines/métabolisme , Tumeurs/anatomopathologie , Polyosides/métabolisme , Plaquettes/métabolisme , Glycosylation , Métastase tumorale/anatomopathologie , Microenvironnement tumoral
7.
Int J Biol Sci ; 20(3): 1110-1124, 2024.
Article de Anglais | MEDLINE | ID: mdl-38322116

RÉSUMÉ

At present, tumor metastasis still remains the leading contributor to high recurrence and mortality in cancer patients. There have been no clinically effective therapeutic strategies for treating patients with metastatic cancer. In recent years, a growing body of evidence has shown that the pre-metastatic niche (PMN) plays a crucial role in driving tumor metastasis. Nevertheless, a clear and detailed understanding of the formation of PMN is still lacking given the fact that PMN formation involves in a wealth of complicated communications and underlying mechanisms between primary tumors and metastatic target organs. Despite that the roles of numerous components including tumor exosomes and extracellular vesicles in influencing the evolution of PMN have been well documented, the involvement of cancer-associated fibroblasts (CAFs) in the tumor microenvironment for controlling PMN formation is frequently overlooked. It has been increasingly recognized that fibroblasts trigger the formation of PMN by virtue of modulating exosomes, metabolism and so on. In this review, we mainly summarize the underlying mechanisms of fibroblasts from diverse origins in exerting impacts on PMN evolution, and further highlight the prospective strategies for targeting fibroblasts to prevent PMN formation.


Sujet(s)
Fibroblastes associés au cancer , Exosomes , Vésicules extracellulaires , Tumeurs , Humains , Études prospectives , Tumeurs/métabolisme , Exosomes/métabolisme , Vésicules extracellulaires/métabolisme , Fibroblastes/métabolisme , Fibroblastes associés au cancer/métabolisme , Microenvironnement tumoral , Métastase tumorale/anatomopathologie
8.
Nat Commun ; 15(1): 1362, 2024 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-38355937

RÉSUMÉ

Metastasis is the major cause of lung cancer-related death, but the mechanisms governing lung tumor metastasis remain incompletely elucidated. SE translocation (SET) is overexpressed in lung tumors and correlates with unfavorable prognosis. Here we uncover SET-associated transcription factor, zinc finger and BTB domain-containing protein 11 (ZBTB11), as a prometastatic regulator in lung tumors. SET interacts and collaborates with ZBTB11 to promote lung cancer cell migration and invasion, primarily through SET-ZBTB11 complex-mediated transcriptional activation of matrix metalloproteinase-9 (MMP9). Additionally, by transcriptional repression of proline-rich Gla protein 2 (PRRG2), ZBTB11 links Yes-associated protein 1 (YAP1) activation to drive lung tumor metastasis independently of SET-ZBTB11 complex. Loss of ZBTB11 suppresses distal metastasis in a lung tumor mouse model. Overexpression of ZBTB11 is recapitulated in human metastatic lung tumors and correlates with diminished survival. Our study demonstrates ZBTB11 as a key metastatic regulator and reveals diverse mechanisms by which ZBTB11 modulates lung tumor metastasis.


Sujet(s)
Tumeurs du poumon , Animaux , Humains , Souris , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation de l'expression des gènes , Poumon/anatomopathologie , Tumeurs du poumon/anatomopathologie , Invasion tumorale/anatomopathologie , Métastase tumorale/anatomopathologie , Protéines oncogènes/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
9.
Mol Cancer ; 23(1): 18, 2024 01 19.
Article de Anglais | MEDLINE | ID: mdl-38243280

RÉSUMÉ

The production and release of tumor-derived small extracellular vesicles (TDSEVs) from cancerous cells play a pivotal role in the propagation of cancer, through genetic and biological communication with healthy cells. TDSEVs are known to orchestrate the invasion-metastasis cascade via diverse pathways. Regulation of early metastasis processes, pre-metastatic niche formation, immune system regulation, angiogenesis initiation, extracellular matrix (ECM) remodeling, immune modulation, and epithelial-mesenchymal transition (EMT) are among the pathways regulated by TDSEVs. MicroRNAs (miRs) carried within TDSEVs play a pivotal role as a double-edged sword and can either promote metastasis or inhibit cancer progression. TDSEVs can serve as excellent markers for early detection of tumors, and tumor metastases. From a therapeutic point of view, the risk of cancer metastasis may be reduced by limiting the production of TDSEVs from tumor cells. On the other hand, TDSEVs represent a promising approach for in vivo delivery of therapeutic cargo to tumor cells. The present review article discusses the recent developments and the current views of TDSEVs in the field of cancer research and clinical applications.


Sujet(s)
Vésicules extracellulaires , microARN , Tumeurs , Humains , Pertinence clinique , Tumeurs/anatomopathologie , microARN/génétique , Communication cellulaire , Transition épithélio-mésenchymateuse , Microenvironnement tumoral , Métastase tumorale/anatomopathologie
10.
Biomed Pharmacother ; 171: 116108, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38218079

RÉSUMÉ

Metastasis is the leading cause of cancer-related deaths. Despite this relevance, there is no specific therapy targeting metastasis. The interaction of the tumor cell with platelets, forming microemboli is crucial for successful hematogenous dissemination. Heparin disrupts it by a P-selectin-mediated event. However, its clinical use for this purpose is hindered by the requirement of high doses, leading to anticoagulant-related side effects. In this study, we obtained a low-anticoagulant heparin through the fractionation of a pharmaceutical bovine heparin. This derivative was referred to as LA-hep and we investigated its efficacy in inhibiting metastases and explored its capacity of suppressing the interaction between tumor cells and platelets. Our data revealed that LA-hep is as efficient as porcine unfractionated heparin in attenuating lung metastases from melanoma and colon adenocarcinoma cells in an assay with a single intravenous administration. It also prevents platelet arrest shortly after cell injection in wild-type mice and suppresses melanoma-platelets interaction in vitro. Moreover, LA-hep blocks P-selectin's direct binding to tumor cells and platelet aggregation, providing further evidence for the role of P-selectin as a molecular target. Even in P-selectin-depleted mice which developed a reduced number of metastatic foci, both porcine heparin and LA-hep further inhibited metastasis burden. This suggests evidence of an additional mechanism of antimetastatic action. Therefore, our results indicate a dissociation between the heparin anticoagulant and antimetastatic effects. Considering the simple and highly reproducible methodology used to purify LA-hep along with the data presented here, LA-hep emerges as a promising drug for future use in preventing metastasis in cancer patients.


Sujet(s)
Adénocarcinome , Tumeurs du côlon , Mélanome , Humains , Animaux , Bovins , Souris , Héparine/pharmacologie , Anticoagulants/pharmacologie , Sélectine P/métabolisme , Mélanome/anatomopathologie , Adénocarcinome/anatomopathologie , Tumeurs du côlon/anatomopathologie , Plaquettes/métabolisme , Préparations pharmaceutiques/métabolisme , Métastase tumorale/anatomopathologie
11.
Histol Histopathol ; 39(2): 145-152, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37458321

RÉSUMÉ

Platelets are generally considered as the main functional unit of the coagulation system. However, more and more studies have confirmed that platelets also have an important relationship with tumor progression. Tumor cells can utilize platelets to promote their own infiltration and hematogenous metastasis, and platelets are activated and aggregated in this process. Therefore, platelet aggregation may be a concomitant marker of tumor progression. This is of great significance for predicting tumor metastasis before timely treatments.


Sujet(s)
Plaquettes , Agrégation plaquettaire , Humains , Métastase tumorale/anatomopathologie
12.
Scand J Gastroenterol ; 59(3): 354-360, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38042983

RÉSUMÉ

BACKGROUND: Pancreatic metastases from renal cell carcinoma (RCC) are rare. This study evaluated the surgical pathology and outcomes after resection of RCC metastases to the pancreas. MATERIAL AND METHODS: A retrospective review of from 1 January 2011 to 31 December 2021, of patients who underwent pancreatic surgery for metastases from RCC. Data were retrieved from a prospectively managed database and patient demographics, comorbidities, pathology, perioperative outcomes, and overall survival were analyzed. Median overall survival (OS) and disease-free survival (DFS) were estimated by the Kaplan-Meier method. RESULTS: There were 25 patients (17 males, 8 females, median age 66 range 51 - 79 year), all with metachronous metastases. Median time from resection of the primary to operation for pancreatic RCC was 95.6 (12.0 - 309.7) months. Twenty-four patients were operated with intended cure (four pancreaticoduodenectomies, three total pancreatectomies, 17 distal pancreatectomies) and one patient had abortive surgery due to dissemination. Postoperative surgical complications occurred in nine patients (36%), and one patient died during hospital stay. Eight patients (33.3%) developed exocrine and/or endocrine insufficiency after pancreatic resection. Fifteen patients (60%) had recurrence 21.7 (4.9 - 61.6) months after pancreatic operation. Five patients (25%) died from RCC during follow-up 46.3 (25.6 - 134.8) months after pancreatic resection. Five-year OS and DFS were83.6% and 32.3%, respectively. Median OS after pancreatic surgery was 134.8 months, independent of resection of previous extrapancreatic metastases. CONCLUSIONS: Pancreatic resection for metastases from RCC offers favorable prognosis with a curative potential and should be considered a valuable treatment option even in the era of novel targeted treatment.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Pancréatectomie , Tumeurs du pancréas , Sujet âgé , Femelle , Humains , Mâle , Néphrocarcinome/chirurgie , Néphrocarcinome/secondaire , Tumeurs du rein/chirurgie , Tumeurs du rein/anatomopathologie , Pancréas/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/chirurgie , Pronostic , Études rétrospectives , Métastase tumorale/anatomopathologie , Complications postopératoires
13.
Semin Thromb Hemost ; 50(3): 455-461, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37832586

RÉSUMÉ

Alongside their conventional roles in thrombosis and hemostasis, platelets have long been associated with nonhemostatic pathologies, including tumor cell metastasis. Numerous mechanistic studies have since demonstrated that the direct binding of platelets to intravascular tumor cells promotes key hallmarks of metastasis, including survival in circulation and tumor cell arrest at secondary sites. However, platelets also interact with nonmalignant cells that make up the stromal and immune compartments within both primary and metastatic tumors. This review will first provide a brief historical perspective on platelet contributions to metastatic disease before discussing the emerging roles that platelets play in creating microenvironments that likely support successful tumor cell metastasis.


Sujet(s)
Tumeurs , Microenvironnement tumoral , Humains , Plaquettes/métabolisme , Tumeurs/métabolisme , Hémostase , Métastase tumorale/anatomopathologie
14.
Cell ; 186(26): 5719-5738.e28, 2023 12 21.
Article de Anglais | MEDLINE | ID: mdl-38056463

RÉSUMÉ

Tumor-associated hydrocephalus (TAH) is a common and lethal complication of brain metastases. Although other factors beyond mechanical obstructions have been suggested, the exact mechanisms are unknown. Using single-nucleus RNA sequencing and spatial transcriptomics, we find that a distinct population of mast cells locate in the choroid plexus and dramatically increase during TAH. Genetic fate tracing and intracranial mast-cell-specific tryptase knockout showed that choroid plexus mast cells (CPMCs) disrupt cilia of choroid plexus epithelia via the tryptase-PAR2-FoxJ1 pathway and consequently increase cerebrospinal fluid production. Mast cells are also found in the human choroid plexus. Levels of tryptase in cerebrospinal fluid are closely associated with clinical severity of TAH. BMS-262084, an inhibitor of tryptase, can cross the blood-brain barrier, inhibit TAH in vivo, and alleviate mast-cell-induced damage of epithelial cilia in a human pluripotent stem-cell-derived choroid plexus organoid model. Collectively, we uncover the function of CPMCs and provide an attractive therapy for TAH.


Sujet(s)
Tumeurs du cerveau , Plexus choroïde , Hydrocéphalie , Mastocytes , Humains , Tumeurs du cerveau/secondaire , Plexus choroïde/métabolisme , Plexus choroïde/anatomopathologie , Hydrocéphalie/métabolisme , Hydrocéphalie/anatomopathologie , Mastocytes/métabolisme , Mastocytes/anatomopathologie , Tryptases/liquide cérébrospinal , Métastase tumorale/anatomopathologie
15.
Mol Cancer ; 22(1): 193, 2023 11 30.
Article de Anglais | MEDLINE | ID: mdl-38037077

RÉSUMÉ

Current research has demonstrated that extracellular vesicles (EVs) and circulating tumor cells (CTCs) are very closely related in the process of distant tumor metastasis. Primary tumors are shed and released into the bloodstream to form CTCs that are referred to as seeds to colonize and grow in soil-like distant target organs, while EVs of tumor and nontumor origin act as fertilizers in the process of tumor metastasis. There is no previous text that provides a comprehensive review of the role of EVs on CTCs during tumor metastasis. In this paper, we reviewed the mechanisms of EVs on CTCs during tumor metastasis, including the ability of EVs to enhance the shedding of CTCs, protect CTCs in circulation and determine the direction of CTC metastasis, thus affecting the distant metastasis of tumors.


Sujet(s)
Vésicules extracellulaires , Cellules tumorales circulantes , Humains , Cellules tumorales circulantes/anatomopathologie , Métastase tumorale/anatomopathologie , Marqueurs biologiques tumoraux
16.
Int J Mol Sci ; 24(21)2023 Oct 26.
Article de Anglais | MEDLINE | ID: mdl-37958607

RÉSUMÉ

Breast cancer (BC) remains one of the most common malignancies among women worldwide. Breast cancer shows metastatic heterogeneity with priority to different organs, which leads to differences in prognosis and response to therapy among patients. The main targets for metastasis in BC are the bone, lung, liver and brain. The molecular mechanism of BC organ-specificity is still under investigation. In recent years, the appearance of new genomic approaches has led to unprecedented changes in the understanding of breast cancer metastasis organ-specificity and has provided a new platform for the development of more effective therapeutic agents. This review summarises recent data on molecular organ-specific markers of metastasis as the basis of a possible therapeutic approach in order to improve the diagnosis and prognosis of patients with metastatically heterogeneous breast cancer.


Sujet(s)
Tumeurs du sein , Tumeurs du poumon , Mélanome , Seconde tumeur primitive , Femelle , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du poumon/génétique , Poumon/anatomopathologie , Mélanome/anatomopathologie , Seconde tumeur primitive/anatomopathologie , Métastase tumorale/anatomopathologie ,
17.
Life Sci ; 335: 122255, 2023 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-37967792

RÉSUMÉ

BACKGROUND: Cancer metastasis is a major cause of cancer-related deaths, emphasizing the urgent need for effective therapies. Although it has been shown that GMI, a fungal protein from Ganoderma microsporum, could suppress primary tumor growth in a wide spectrum of cancer types, it is still unclear whether GMI exhibits anti-metastasis properties, particularly in lung cancers. Further investigation is needed. AIMS AND OBJECTIVES: The objective of this study is to investigate the potential inhibitory effects of GMI on lung cancer metastasis in vivo. Utilizing systematic and comprehensive approaches, our research aims to elucidate the underlying molecular mechanisms responsible for the anti-metastatic effects. MATERIALS AND METHODS: In vitro migration and cell adhesion assays addressed the epithelial-to-mesenchymal transition (EMT)-related phenotype. Proteomic and bioinformatic analyses identified the GMI-regulated proteins and cellular responses. GMI-treated LLC1-bearing mice were analyzed using IVIS Spectrum to assess the anti-metastatic effect. KEY FINDINGS: GMI inhibits EMT as well as cell migration. GMI disrupts cell adhesion and downregulates integrin, resulting in inhibition of phosphorylated FAK. GMI induces macropinocytosis and lysosome-mediated degradation of integrin αv, α5, α6 and ß1. GMI downregulates Slug via inhibition of FAK activity, which in turn enhances expressions of epithelial-related markers and decreases cell mobility. Mechanistically, GMI-induced FAK inhibition engenders MDM2 expression and enhances MDM2/p21/Slug complex formation, leading to Slug degradation. GMI treatment reduces the metastatic pulmonary lesion and prolongs the survival of LLC1-bearing mice. SIGNIFICANCE: Our findings highlight GMI as a promising therapeutic candidate for metastatic lung cancers, offering potential avenues for further research and drug development.


Sujet(s)
Tumeurs du poumon , Animaux , Souris , Tumeurs du poumon/anatomopathologie , Contacts focaux/métabolisme , Contacts focaux/anatomopathologie , Protéomique , Lignée cellulaire tumorale , Mouvement cellulaire , Transition épithélio-mésenchymateuse , Métastase tumorale/anatomopathologie
18.
Cell Rep ; 42(12): 113470, 2023 12 26.
Article de Anglais | MEDLINE | ID: mdl-37979166

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) empowers epithelial cells with mesenchymal and stem-like attributes, facilitating metastasis, a leading cause of cancer-related mortality. Hybrid epithelial-mesenchymal (E/M) cells, retaining both epithelial and mesenchymal traits, exhibit heightened metastatic potential and stemness. The mesenchymal intermediate filament, vimentin, is upregulated during EMT, enhancing the resilience and invasiveness of carcinoma cells. The phosphorylation of vimentin is critical to its structure and function. Here, we identify that stabilizing vimentin phosphorylation at serine 56 induces multinucleation, specifically in hybrid E/M cells with stemness properties but not epithelial or mesenchymal cells. Cancer stem-like cells are especially susceptible to vimentin-induced multinucleation relative to differentiated cells, leading to a reduction in self-renewal and stemness. As a result, vimentin-induced multinucleation leads to sustained inhibition of stemness properties, tumor initiation, and metastasis. These observations indicate that a single, targetable phosphorylation event in vimentin is critical for stemness and metastasis in carcinomas with hybrid E/M properties.


Sujet(s)
Carcinomes , Filaments intermédiaires , Humains , Vimentine/métabolisme , Phosphorylation , Filaments intermédiaires/métabolisme , Filaments intermédiaires/anatomopathologie , Carcinomes/anatomopathologie , Cellules épithéliales/métabolisme , Transition épithélio-mésenchymateuse , Cellules souches tumorales/métabolisme , Lignée cellulaire tumorale , Métastase tumorale/anatomopathologie
19.
Animal Model Exp Med ; 6(5): 399-408, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37902101

RÉSUMÉ

The premetastatic niches (PMN) formed by primary tumor-derived molecules regulate distant organs and tissues to further favor tumor colonization. Targeted PMN therapy may prevent tumor metastasis in the early stages, which is becoming increasingly important. At present, there is a lack of in-depth understanding of the cellular and molecular characteristics of the PMN. Here, we summarize current research advances on the cellular and molecular characteristics of the PMN. We emphasize that PMN intervention is a potential therapeutic strategy for early prevention of tumor metastasis, which provides a promising basis for future research and clinical application.


Sujet(s)
Métastase tumorale , Tumeurs , Humains , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/thérapie , Métastase tumorale/anatomopathologie
20.
Cancer Lett ; 578: 216438, 2023 12 01.
Article de Anglais | MEDLINE | ID: mdl-37827326

RÉSUMÉ

Metastasis is the primary cause of death in colorectal cancer (CRC). Thyroid hormone receptor interacting protein 6 (TRIP6) is an adaptor protein that regulates cell motility. Here, we aim to elucidate the role of TRIP6 in driving CRC tumorigenesis and metastasis and evaluate its potential as a therapeutic target. TRIP6 mRNA is up-regulated in CRC compared to adjacent normal tissues in three independent cohorts (all P < 0.0001), especially in liver metastases (P < 0.001). High TRIP6 expression predicts poor prognosis of CRC patients in our cohort (P = 0.01) and TCGA cohort (P = 0.02). Colon-specific TRIP6 overexpression (Trip6KIVillin-Cre) in mice accelerated azoxymethane (AOM)-induced CRC (P < 0.05) and submucosal invasion (P < 0.0001). In contrast, TRIP6 knockout (Trip6+/- mice) slowed tumorigenesis (P < 0.05). Consistently, TRIP6 overexpression in CRC cells promoted epithelial-mesenchymal transition (EMT), cell migration/invasion in vitro, and metastases in vivo (all P < 0.05), whereas knockdown of TRIP6 exerted opposite phenotypes. Mechanistically, TRIP6 interacted PDZ domain-containing proteins such as PARD3 to impair tight junctions, evidenced by decreased tight junction markers and gut permeability dysfunction, inhibit PTEN, and activate oncogenic Akt signaling. TRIP6-induced pro-metastatic phenotypes and Akt activation depends on PARD3. Targeting TRIP6 by VNP-encapsulated TRIP6-siRNA synergized with Oxaliplatin and 5-Fluorouracil to suppress CRC liver metastases. In conclusion, TRIP6 promotes CRC metastasis by directly interacting with PARD3 to disrupt tight junctions and activating Akt signaling. Targeting of TRIP6 in combination with chemotherapy is a promising strategy for the treatment of metastatic CRC.


Sujet(s)
Tumeurs colorectales , Tumeurs du foie , Animaux , Humains , Souris , Carcinogenèse , Lignée cellulaire tumorale , Mouvement cellulaire , Transformation cellulaire néoplasique/génétique , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Résistance aux substances , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Métastase tumorale/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Jonctions serrées/métabolisme , Jonctions serrées/anatomopathologie , Facteurs de transcription/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...