Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 40(18): e108647, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34459003

RESUMO

The process of epithelial-mesenchymal transition (EMT) is fundamental for embryonic morphogenesis. Cells undergoing it lose epithelial characteristics and integrity, acquire mesenchymal features, and become motile. In cancer, this program is hijacked to confer essential changes in morphology and motility that fuel invasion. In addition, EMT is increasingly understood to orchestrate a large variety of complementary cancer features, such as tumor cell stemness, tumorigenicity, resistance to therapy and adaptation to changes in the microenvironment. In this review, we summarize recent findings related to these various classical and non-classical functions, and introduce EMT as a true tumorigenic multi-tool, involved in many aspects of cancer. We suggest that therapeutic targeting of the EMT process will-if acknowledging these complexities-be a possibility to concurrently interfere with tumor progression on many levels.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias/etiologia , Neoplasias/patologia , Microambiente Tumoral , Animais , Biomarcadores , Transformação Celular Neoplásica , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/metabolismo , Transdução de Sinais , Microambiente Tumoral/genética
2.
J Pathol ; 262(3): 347-361, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38235615

RESUMO

Partial epithelial-mesenchymal transition (p-EMT) has recently been identified as a hybrid state consisting of cells with both epithelial and mesenchymal characteristics and is associated with the migration, metastasis, and chemoresistance of cancer cells. Here, we describe the induction of p-EMT in starved colorectal cancer (CRC) cells and identify a p-EMT gene signature that can predict prognosis. Functional characterisation of starvation-induced p-EMT in HCT116, DLD1, and HT29 cells showed changes in proliferation, morphology, and drug sensitivity, supported by in vivo studies using the chorioallantoic membrane model. An EMT-specific quantitative polymerase chain reaction (qPCR) array was used to screen for deregulated genes, leading to the establishment of an in silico gene signature that was correlated with poor disease-free survival in CRC patients along with the CRC consensus molecular subtype CMS4. Among the significantly deregulated p-EMT genes, a triple-gene signature consisting of SERPINE1, SOX10, and epidermal growth factor receptor (EGFR) was identified. Starvation-induced p-EMT was characterised by increased migratory potential and chemoresistance, as well as E-cadherin processing and internalisation. Both gene signature and E-cadherin alterations could be reversed by the proteasomal inhibitor MG132. Spatially resolving EGFR expression with high-resolution immunofluorescence imaging identified a proliferation stop in starved CRC cells caused by EGFR internalisation. In conclusion, we have gained insight into a previously undiscovered EMT mechanism that may become relevant when tumour cells are under nutrient stress, as seen in early stages of metastasis. Targeting this process of tumour cell dissemination might help to prevent EMT and overcome drug resistance. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/patologia , Proliferação de Células , Transição Epitelial-Mesenquimal/genética , Receptores ErbB , Linhagem Celular Tumoral , Caderinas/genética , Caderinas/metabolismo , Movimento Celular
3.
Semin Cancer Biol ; 97: 1-11, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37944215

RESUMO

Transforming growth factor-ß (TGF-ß) signaling regulates cell-specific programs involved in embryonic development, wound-healing, and immune homeostasis. Yet, during tumor progression, these TGF-ß-mediated programs are altered, leading to epithelial cell plasticity and a reprogramming of epithelial cells into mesenchymal lineages through epithelial-to-mesenchymal transition (EMT), a critical developmental program in morphogenesis and organogenesis. These changes, in turn, lead to enhanced carcinoma cell invasion, metastasis, immune cell differentiation, immune evasion, and chemotherapy resistance. Here, we discuss EMT as one of the critical programs associated with carcinoma cell plasticity and the influence exerted by TGF-ß on carcinoma status and function. We further explore the composition of carcinoma and other cell populations within the tumor microenvironment, and consider the relevant outcomes related to the programs associated with cancer treatment resistance.


Assuntos
Carcinoma , Fator de Crescimento Transformador beta , Humanos , Transição Epitelial-Mesenquimal/genética , Células Epiteliais , Transdução de Sinais , Linhagem Celular Tumoral , Microambiente Tumoral
4.
J Transl Med ; 21(1): 267, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37076857

RESUMO

BACKGROUND: Oral squamous cell carcinoma (OSCC), an HPV-negative head and neck cancer, frequently metastasizes to the regional lymph nodes but only occasionally beyond. Initial phases of metastasis are associated with an epithelial-mesenchymal transition (EMT), while the consolidation phase is associated with mesenchymal-epithelial transition (MET). This dynamic is referred to as epithelial-mesenchymal plasticity (EMP). While it is known that EMP is essential for cancer cell invasion and metastatic spread, less is known about the heterogeneity of EMP states and even less about the heterogeneity between primary and metastatic lesions. METHODS: To assess both the heterogeneity of EMP states in OSCC cells and their effects on stromal cells, we performed single-cell RNA sequencing (scRNAseq) of 5 primary tumors, 9 matching metastatic and 5 tumor-free lymph nodes and re-analyzed publicly available scRNAseq data of 9 additional primary tumors. For examining the cell type composition, we performed bulk transcriptome sequencing. Protein expression of selected genes were confirmed by immunohistochemistry. RESULTS: From the 23 OSCC lesions, the single cell transcriptomes of a total of 7263 carcinoma cells were available for in-depth analyses. We initially focused on one lesion to avoid confounding inter-patient heterogeneity and identified OSCC cells expressing genes characteristic of different epithelial and partial EMT stages. RNA velocity and the increase in inferred copy number variations indicated a progressive trajectory towards epithelial differentiation in this metastatic lesion, i.e., cells likely underwent MET. Extension to all samples revealed a less stringent but essentially similar pattern. Interestingly, MET cells show increased activity of the EMT-activator ZEB1. Immunohistochemistry confirmed that ZEB1 was co-expressed with the epithelial marker cornifin B in individual tumor cells. The lack of E-cadherin mRNA expression suggests this is a partial MET. Within the tumor microenvironment we found immunomodulating fibroblasts that were maintained in primary and metastatic OSCC. CONCLUSIONS: This study reveals that EMP enables different partial EMT and epithelial phenotypes of OSCC cells, which are endowed with capabilities essential for the different stages of the metastatic process, including maintenance of cellular integrity. During MET, ZEB1 appears to be functionally active, indicating a more complex role of ZEB1 than mere induction of EMT.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Metástase Linfática , Variações do Número de Cópias de DNA , Caderinas/genética , Diferenciação Celular , Transição Epitelial-Mesenquimal/genética , Linhagem Celular Tumoral , Microambiente Tumoral , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
5.
EMBO Rep ; 22(9): e51872, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34324787

RESUMO

Epithelial plasticity, or epithelial-to-mesenchymal transition (EMT), is a well-recognized form of cellular plasticity, which endows tumor cells with invasive properties and alters their sensitivity to various agents, thus representing a major challenge to cancer therapy. It is increasingly accepted that carcinoma cells exist along a continuum of hybrid epithelial-mesenchymal (E-M) states and that cells exhibiting such partial EMT (P-EMT) states have greater metastatic competence than those characterized by either extreme (E or M). We described recently a P-EMT program operating in vivo by which carcinoma cells lose their epithelial state through post-translational programs. Here, we investigate the underlying mechanisms and report that prolonged calcium signaling induces a P-EMT characterized by the internalization of membrane-associated E-cadherin (ECAD) and other epithelial proteins as well as an increase in cellular migration and invasion. Signaling through Gαq-associated G-protein-coupled receptors (GPCRs) recapitulates these effects, which operate through the downstream activation of calmodulin-Camk2b signaling. These results implicate calcium signaling as a trigger for the acquisition of hybrid/partial epithelial-mesenchymal states in carcinoma cells.


Assuntos
Sinalização do Cálcio , Transição Epitelial-Mesenquimal , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Plasticidade Celular
6.
Int J Mol Sci ; 24(3)2023 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-36768876

RESUMO

Cancer stem cells (CSCs) have been identified and characterized in both hematopoietic and solid tumors. Their existence was first predicted by Virchow and Cohnheim in the 1870s. Later, many studies showed that CSCs can be identified and isolated by their expression of specific cell markers. The significance of CSCs with respect to tumor biology and anti-cancer treatment lies in their ability to maintain quiescence with very slow proliferation, indefinite self-renewal, differentiation, and trans-differentiation such as epithelial-mesenchymal transition (EMT) and its reverse process mesenchymal-epithelial transition (MET). The ability for detachment, migration, extra- and intravasation, invasion and thereby of completing all necessary steps of the metastatic cascade highlights their significance for metastasis. CSCs comprise the cancer cell populations responsible for tumor growth, resistance to therapies and cancer metastasis. In this review, the history of the CSC theory, their identification and characterization and their biology are described. The contribution of the CSC ability to undergo EMT for cancer metastasis is discussed. Recently, novel strategies for drug development have focused on the elimination of the CSCs specifically. The unique functional and molecular properties of CSCs are discussed as possible therapeutic vulnerabilities for the development of novel anti-metastasis treatments. Prospectively, this may provide precise personalized anti-cancer treatments with improved therapeutic efficiency with fewer side effects and leading to better prognosis.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias , Humanos , Neoplasias/metabolismo , Diferenciação Celular , Células-Tronco Neoplásicas/metabolismo
7.
Phys Biol ; 19(2)2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-34986465

RESUMO

The epithelial-mesenchymal transition (EMT) is a biological phenomenon associated with explicit phenotypic and molecular changes in cellular traits. Unlike the earlier-held popular belief of it being a binary process, EMT is now thought of as a landscape including diverse hybrid E/M phenotypes manifested by varying degrees of the transition. These hybrid cells can co-express both epithelial and mesenchymal markers and/or functional traits, and can possess the property of collective cell migration, enhanced tumor-initiating ability, and immune/targeted therapy-evasive features, all of which are often associated with worse patient outcomes. These characteristics of the hybrid E/M cells have led to a surge in studies that map their biophysical and biochemical hallmarks that can be helpful in exploiting their therapeutic vulnerabilities. This review discusses recent advances made in investigating hybrid E/M phenotype(s) from diverse biophysical and biochemical aspects by integrating live cell-imaging, cellular morphology quantification and mathematical modeling, and highlights a set of questions that remain unanswered about the dynamics of hybrid E/M states.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias , Biofísica , Movimento Celular , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Fenótipo
8.
Endocr Res ; 47(2): 64-79, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35168462

RESUMO

BACKGROUND: Epithelial-mesenchymal transition (EMT) of tumor cells is a prerequisite to cancer cell invasion and metastasis. This process involves a network of molecular alterations. Androgen receptor (AR) plays an important role in the biology of breast cancers, particularly those dependent on AR expression like luminal AR (LAR) breast cancer subtype. We have recently reported that the AR agonist, dihydrotestosterone (DHT), induces a mesenchymal transition of MDA-MB-453 cells, concomitant with transcriptional up-regulation of Slug and regulator of G protein signaling 2 (RGS2). OBJECTIVE: The role of Slug and RGS2 in mediating the DHT-induced effects in these cells was investigated. METHODS: MDA-MB-453 cells were used as a model system of LAR breast cancer. Immunofluorescence was used to examine cell morphology and protein localization. Protein expression was analyzed by immunoblotting. Protein localization was confirmed by cell fractionation followed by immunoblotting. Protein-protein interaction was confirmed by co-immunoprecipitation followed by immunoblotting. Transwell membranes were used to assess cell migration. Transfection of cells with siRNA molecules that target Slug and RGS2 mRNA was utilized to delineate the modes of action of these two molecules. RESULTS: Treatment of MDA-MB-453 cells with DHT induced the expression of both proteins. In addition, AR-Slug, AR-RGS2, and Slug-RGS2 interactions were observed shortly after AR activation. Knocking down Slug abrogated the basal, but not the DHT-induced, cell migration and blocked DHT-induced mesenchymal transition. On the other hand, RGS2 knocked-down cells had an increased level of Slug protein and assumed mesenchymal cell morphology with induced migration, and the addition of DHT further elongated cell morphology and stimulated their migration. Inhibition of AR or ß-catenin reverted the RGS2 knocked-down cells to the epithelial phenotype, but only inhibition of AR blocked their DHT-induced migration. CONCLUSIONS: These results suggest the involvement of RGS2 and Slug in a complex molecular network regulating the DHT-induced mesenchymal features in MDA-MB-453 cells. The study may offer a better understanding of the biological role of AR in breast cancer toward devising AR-based therapeutic strategies.


Assuntos
Neoplasias da Mama , Proteínas RGS , Androgênios/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Di-Hidrotestosterona/farmacologia , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Humanos , Fatores de Transcrição da Família Snail
9.
Int J Mol Sci ; 24(1)2022 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-36613756

RESUMO

Whole exome sequencing of invasive mammary carcinomas revealed the association of mutations in PTEN and ZFHX3 tumor suppressor genes (TSGs). We generated single and combined PTEN and ZFHX3 knock-outs (KOs) in the immortalized mammary epithelial cell line MCF10A to study the role of these genes and their potential synergy in migration regulation. Inactivation of PTEN, but not ZFHX3, induced the formation of large colonies in soft agar. ZFHX3 inactivation in PTEN KO, however, increased colony numbers and normalized their size. Cell migration was affected in different ways upon PTEN and ZFHX3 KO. Inactivation of PTEN enhanced coordinated cell motility and thus, the collective migration of epithelial islets and wound healing. In contrast, ZFHX3 knockout resulted in the acquisition of uncoordinated cell movement associated with the appearance of immature adhesive junctions (AJs) and the increased expression of the mesenchymal marker vimentin. Inactivation of the two TSGs thus induces different stages of partial epithelial-to-mesenchymal transitions (EMT). Upon double KO (DKO), cells displayed still another motile state, characterized by a decreased coordination in collective migration and high levels of vimentin but a restoration of mature linear AJs. This study illustrates the plasticity of migration modes of mammary cells transformed by a combination of cancer-associated genes.


Assuntos
Mama , Células Epiteliais , Humanos , Vimentina/metabolismo , Mama/metabolismo , Células Epiteliais/metabolismo , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Linhagem Celular Tumoral , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas de Homeodomínio/genética
10.
Endocr Res ; 46(3): 114-128, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33703980

RESUMO

Purpose The cellular and molecular dynamics of DHT-induced EMT in MDA-MB-453 cells were investigated.Methods:PCR arrays were used to examine the expression of EMT-regulatory genes. Immunoblotting was used to detect protein levels and confirm protein-protein interaction following immunoprecipitation. Immunofluorescence was used to observe rearrangement of the actin cytoskeleton and cell morphology. Cell migration was assessed by transwell assayResults: Change of cell morphology was concomitant with increased cell migration after treating cells with DHT. Exposure of cells to DHT for one hour was sufficient to induce changes in cell morphology and actin cytoskeleton after 72 hours indicating altered gene expression. A long-term lasting nuclear translocation of AR was observed after a short exposure of cells to DHT. Investigating the expression of 84 EMT-related genes revealed down-expression of ß-catenin, N-cadherin, and TCF-4 and increased expression of Slug, all of which were confirmed at the protein level. Yet, not only early interaction of AR and ß-catenin was observed following AR activation, inhibition of ß-catenin blocked DHT-induced mesenchymal transition and migration. Wnt signaling was found to be partially important in DHT-induced morphological alteration. The mesenchymal transition of cells could be induced by treating cells with an inhibitor of glycogen synthase kinase-3ß, an enzyme that inhibits ß-catenin; this morphological transition could be reversed by antagonizing AR suggesting that AR functions downstream of ß-catenin.Conclusions: These results suggest that MDA-MB-453 cells undergo partial EMT induced by DHT, ß-catenin is critical for this phenotypic change, and AR probably reciprocally mediates the mesenchymal transition of these cells upon activation of GSK-3 ß.


Assuntos
Neoplasias da Mama/metabolismo , Di-Hidrotestosterona/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Receptores Androgênicos/metabolismo , beta Catenina/metabolismo , Linhagem Celular Tumoral , Humanos
11.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34884649

RESUMO

Prostate cancer is the most common cancer in American men and the second leading cause of cancer-related death. Most of these deaths are associated with metastasis, a process involving the epithelial-to-mesenchymal (EMT) transition. Furthermore, growing evidence suggests that partial-EMT (p-EMT) may lead to more aggressive disease than complete EMT. In this study, the EMT-inducing transcription factor Zeb1 was knocked down in mesenchymal PC-3 prostate cancer cells (Zeb1KD) and resulting changes in cellular phenotype were assessed using protein and RNA analysis, invasion and migration assays, cell morphology assays, and DNA methylation chip analysis. Inducible knockdown of Zeb1 resulted in a p-EMT phenotype including co-expression of epithelial and mesenchymal markers, a mixed epithelial/mesenchymal morphology, increased invasion and migration, and enhanced expression of p-EMT markers relative to PC-3 mesenchymal controls (p ≤ 0.05). Treatment of Zeb1KD cells with the global de-methylating drug 5-azacytidine (5-aza) mitigated the observed aggressive p-EMT phenotype (p ≤ 0.05). DNA methylation chip analysis revealed 10 potential targets for identifying and/or targeting aggressive p-EMT prostate cancer in the future. These findings provide a framework to enhance prognostic and/or therapeutic options for aggressive prostate cancer in the future by identifying new p-EMT biomarkers to classify patients with aggressive disease who may benefit from 5-aza treatment.


Assuntos
Metilação de DNA , Neoplasias da Próstata/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/biossíntese , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Conectina/genética , Conectina/metabolismo , Transição Epitelial-Mesenquimal , Humanos , Masculino , Células PC-3 , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
12.
Int J Cancer ; 146(9): 2547-2562, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31506938

RESUMO

Emerging evidence supports the theory that tumor cell clusters efficiently metastasize to distant organs. However, the roles of epithelial-to-mesenchymal transition (EMT) in metastasizing tumor cell clusters have not yet been fully elucidated. To investigate this issue, tumor fragments were dissected from 40 colorectal cancer (CRC) patients and implanted subcutaneously into immunodeficient mice. We observed that tumors developed from the tumor fragments obtained from 28 of the 40 CRC patients. The tumors were then dissociated into cell suspensions to be orthotopically injected into secondary mice. The tumors from 13 of the 28 patients progressed. Furthermore, metastases formed spontaneously in the liver and lungs from the tumor fragments obtained from 8 of these 13 patients. Moreover, employing a mathematical analysis, we showed that tumor cell clusters seeded these metastases significantly more often than did single tumor cells. Membrane E-cadherin- and nuclear ZEB1-positive tumor cells indicating the hybrid epithelial/mesenchymal state were also detected in primary tumors of various CRC patients, and in the corresponding patient-derived xenografts (PDXs) and circulating tumor cell clusters in the bloodstreams of mice. In contrast, ZEB1 staining was barely detectable in the patient-matched liver metastases presumably developing through mesenchymal-to-epithelial transition. Inhibition of E-cadherin or ZEB1 expression by shRNA notably prevented the PDX-derived tumor organoids from colonizing the liver, when injected intrasplenically into mice, indicating E-cadherin and ZEB1 expressions to be required for their metastatic colonization. Taken together, these findings suggest that the epithelial/mesenchymal state mediates metastatic seeding of human CRC cell clusters into distant organs.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/secundário , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Animais , Apoptose , Proliferação de Células , Neoplasias Colorretais/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Metastasis Rev ; 35(4): 645-654, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27878502

RESUMO

Carcinoma cells that are induced to suppress their epithelial features and upregulate mesenchymal gene expression programs acquire traits that promote an invasive and metastatic phenotype. This is achieved through the expression of a program termed the epithelial-to-mesenchymal transition (EMT)-a fundamental cell-biological process that plays key roles in embryogenesis and wound healing. Re-activation of the EMT during cancer promotes disease progression and enhances the metastatic phenotype by bestowing upon previously benign carcinoma cell traits such as migration, invasion, resistance to anoikis, chemoresistance and tumour-initiating potential. Herein, we discuss recent insights into the function of the EMT and cancer cell plasticity during cancer progression, with a focus on their role in promoting successful completion of the later stages of the metastatic cascade.


Assuntos
Neoplasias da Mama/patologia , Plasticidade Celular/fisiologia , Animais , Neoplasias da Mama/tratamento farmacológico , Plasticidade Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Metástase Neoplásica
14.
Proc Natl Acad Sci U S A ; 110(45): 18144-9, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24154725

RESUMO

Forward and backward transitions between epithelial and mesenchymal phenotypes play crucial roles in embryonic development and tissue repair. Aberrantly regulated transitions are also a hallmark of cancer metastasis. The genetic network that regulates these transitions appears to allow for the existence of a hybrid phenotype (epithelial/mesenchymal). Hybrid cells are endowed with mixed epithelial and mesenchymal characteristics, enabling specialized capabilities such as collective cell migration. Cell-fate determination between the three phenotypes is in fact regulated by a circuit composed of two highly interconnected chimeric modules--the miR-34/SNAIL and the miR-200/ZEB mutual-inhibition feedback circuits. Here, we used detailed modeling of microRNA-based regulation to study this core unit. More specifically, we investigated the functions of the two isolated modules and subsequently of the combined unit when the two modules are integrated into the full regulatory circuit. We found that miR-200/ZEB forms a tristable circuit that acts as a ternary switch, driven by miR-34/SNAIL, that is a monostable module that acts as a noise-buffering integrator of internal and external signals. We propose to associate the three stable states--(1,0), (high miR-200)/(low ZEB); (0,1), (low miR-200)/(high ZEB); and (1/2,1/2), (medium miR-200)/(medium ZEB)--with the epithelial, mesenchymal, and hybrid phenotypes, respectively. Our (1/2,1/2) state hypothesis is consistent with recent experimental studies (e.g., ZEB expression measurements in collectively migrating cells) and explains the lack of observed mesenchymal-to-hybrid transitions in metastatic cells and in induced pluripotent stem cells. Testable predictions of dynamic gene expression during complete and partial transitions are presented.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/metabolismo , MicroRNAs/metabolismo , Modelos Biológicos , Fatores de Transcrição/metabolismo , Carcinogênese/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Humanos , Fatores de Transcrição da Família Snail , Homeobox 1 de Ligação a E-box em Dedo de Zinco
15.
Transl Oncol ; 41: 101860, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38262111

RESUMO

INTRODUCTION: Nutrient restriction in cancer cells can activate a number of stress response pathways for cell survival. We aimed to determine mechanistically how nutrient depletion in colorectal cancer (CRC) cells leads to cellular adaptation. MATERIALS AND METHODS: Cell survival under nutrient depletion (ND) was evaluated by colony formation and in vivo tumor formation assays. Lysosomes are activated with ND; therefore, we incubated the ND cells with the V-ATPase inhibitor Bafilomycin A1 (ND+Baf). The expression of epithelial and mesenchymal markers with ND+Baf was determined by RNA sequencing and RT-qPCR while motility was determined with an in vivo Chorioallantoic membrane (CAM) assay. Reorganization of cytoskeletal network and lysosomal positioning was determined by immunocytochemistry. RESULTS: 4 different colorectal cancer (CRC) cell lines under ND showed high viability, tumor forming ability and increased expression of one or more epithelial and mesenchymal markers, suggesting the activation of partial (p)-EMT. We observed a further increase in p-EMT markers, numerous membrane protrusions, decreased cell-cell adhesion in 3D, and increased motility in ND+Baf cells. The protrusions in the ND+Baf cells were primarily mediated by microtubules and enabled the relocalization of lysosomes from the perinuclear region to the periphery. CONCLUSIONS: ND activated p-EMT in CRC cells, which was exacerbated by lysosomal alkalinization. The ND+Baf cells also showed numerous protrusions containing lysosomes, which may lead to lysosomal exocytosis and enhanced motility.

16.
Cancers (Basel) ; 16(2)2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-38254832

RESUMO

BACKGROUND: Expression of programmed death ligand-1 (PD-L1) is related to the prognosis of many solid malignancies, including oral squamous cell carcinoma (OSCC), but the mechanism of PD-L1 induction remains obscure. In this study, we examined the expression of PD-L1 and partial epithelial-mesenchymal transition (pEMT) induced by bacterial lipopolysaccharide (LPS) in OSCC. METHODS: The expression of Toll-like receptor 4 (TLR4) recognizing LPS in OSCC cell lines was analyzed. Moreover, the induction of PD-L1 expression by Porphyromonas gingivalis (P.g) or Escherichia coli (E. coli) LPS and EMT was analyzed by western blotting and RT-PCR. Morphology, proliferation, migration, and invasion capacities were examined upon addition of LPS. PD-L1 within EXOs was examined. RESULTS: PD-L1 expression and pEMT induced by LPS of P.g or E. coli in TLR4-expressing OSCC cell lines were observed. Addition of LPS did not change migration, proliferation, or cell morphology, but increased invasive ability. Moreover, higher expression of PD-L1 was observed in OSCC EXOs with LPS. CONCLUSION: Oral bacterial LPS is involved in enhanced invasive potential in OSCC cells, causing PD-L1 expression and induction of pEMT. The enhancement of PD-L1 expression after addition of LPS may be mediated by EXOs.

17.
Biomed Pharmacother ; 170: 116011, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38157644

RESUMO

Partial epithelial-mesenchymal transition (pEMT) plays a vital role in oral squamous cell carcinoma (OSCC) cervical lymph node metastasis and tumor immune escape as an immune barrier. However, targeted interventions for pEMT have yet to be established. In this study, we generated an αPDPN-Ag2S probe by modifying a Podoplanin(PDPN) monoclonal antibody on the surface of near infrared (NIR)-II Ag2S quantum dots (QDs) with carboxyl groups through an amide reaction. Then, we evaluated its in vivo targeting ability, therapeutic efficacy of eliminating pEMT using αPDPN-Ag2S-mediated NIR-II photoimmunotherapy (PIT) and biological safety. Here, we found that pEMT is related to CD8 + T-cell infiltration in our human OSCC tissue microarray. Compared with PdpnWT SCC7, the slower growth rate of subcutaneous graft tumors implanted with PdpnKD SCC7 was associated with a change in the tumor immune microenvironment (TIM) in an immunocompetent C3H/HeJ mouse model. In vitro, αPDPN-Ag2S plus NIR 808 nm laser irradiation induced SCC7 cell death. In vivo, NIR-II imaging results show that the αPDPN-Ag2S probe has a good active-targeting ability in a 4-nitroquinoline 1-oxide (4NQO)-induced C57 mouse OSCC model and C3H/HeJ SCC7 subcutaneous graft tumor model. Elimination of pEMT cells by NIR-II αPDPN-Ag2S probe-mediated PIT significantly reversed the local immunosuppressive tumor microenvironment and enhanced PD-1 immunotherapy efficacy. The safety profiles of αPDPN-Ag2S in BALB/c mice were also acceptable. Thus, αPDPN-Ag2S has important clinical translational value in predicting the risk of cervical lymph node metastasis. Importantly, our study proposed a new way to improve the efficacy of tumor immunotherapy.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Pontos Quânticos , Camundongos , Animais , Humanos , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Metástase Linfática , Camundongos Endogâmicos C3H , Neoplasias Bucais/terapia , Imunoterapia/métodos , Linhagem Celular Tumoral , Microambiente Tumoral
18.
Sci Rep ; 14(1): 6767, 2024 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-38514830

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is a significant healthcare burden globally. Previous research using single-cell transcriptome analysis identified TGFBI as a crucial marker for the partial-epithelial-mesenchymal transition (partial-EMT) program. However, the precise role of TGFBI in HNSCC progression remains unclear. Therefore, our study aimed to clarify the impact of TGFBI on the malignant behavior of HNSCC cells. Through RNA-sequencing data from the TCGA database, we validated that increased TGFBI expression correlates with a higher occurrence of lymph node metastasis and unfavorable prognosis in HNSCC cases. Functional experiments demonstrated that TGFBI overexpression enhances the ability of sphere formation, indicating stem-cell-like properties. Conversely, TGFBI depletion reduces sphere formation and suppresses the expression of cancer stem cell (CSC) markers. RNA-sequencing analysis of TGFBI-overexpressing and control HNSCC cells revealed TAGLN as a downstream effector mediating TGFBI-induced sphere formation. Remarkably, TAGLN depletion abolished TGFBI-induced sphere formation, while its overexpression rescued the suppressed sphere formation caused by TGFBI depletion. Moreover, elevated TAGLN expression showed correlations with the expression of TGFBI and partial-EMT-related genes in HNSCC cases. In conclusion, our findings suggest that TGFBI may promote CSC properties through the upregulation of TAGLN. These novel insights shed light on the involvement of the TGFBI-TAGLN axis in HNSCC progression and hold implications for the development of targeted therapies.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/patologia , Linhagem Celular Tumoral , Células-Tronco Neoplásicas/metabolismo , Transição Epitelial-Mesenquimal/genética , RNA/metabolismo , Regulação Neoplásica da Expressão Gênica
19.
Int Immunopharmacol ; 129: 111650, 2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38342062

RESUMO

Renal fibrosis is a key feature of chronic kidney disease (CKD) progression, whereas no proven effective anti-fibrotic treatments. Forsythiaside A (FTA), derived from Forsythia suspense, has been found to possess nephroprotective properties. However, there is limited research on its anti-fibrotic effects, and its mechanism of action remains unknown. This study aimed to investigate the suppressive effects of FTA on renal fibrosis and explore the underlying mechanisms. In vitro, we established a HK2 cell model induced by transforming growth factor ß1 (TGF-ß1), and in vivo, we used a mice model induced by unilateral ureteral obstruction (UUO). CCK-8 assay, qRT-PCR, Western blotting, immunofluorescence, flow cytometry, histological staining, immunohistochemistry, TUNEL assay, RNA transcriptome sequencing, and molecular docking were performed. The results showed that FTA (40 µM or 80 µM) treatment improved cell viability and suppressed TGF-ß1-induced fibrotic changes and partial epithelial-mesenchymal transition (EMT). Furthermore, FTA treatment reversed the activation of the PI3K/AKT signaling pathway, and THBS1 was identified as the target gene. We found that THBS1 knockdown suppressed the activation of the PI3K/AKT signaling pathway and reduced the fibrosis and partial EMT-related protein level. Conversely, THBS1 overexpression activated the PI3K/AKT signaling pathway and exacerbated renal fibrosis and partial EMT. In vivo, mice were administered FTA (30 or 60 mg/kg) for 2 weeks, and the results demonstrated that FTA administration significantly mitigated tubular injury, tubulointerstitial fibrosis, partial EMT, and apoptosis. In conclusion, FTA inhibited renal fibrosis and partial EMT by targeting THBS1 and inhibiting activation of the PI3K/AKT pathway.


Assuntos
Glicosídeos , Insuficiência Renal Crônica , Obstrução Ureteral , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Simulação de Acoplamento Molecular , Transdução de Sinais , Obstrução Ureteral/tratamento farmacológico , Insuficiência Renal Crônica/patologia , Transição Epitelial-Mesenquimal , Fibrose , Rim/patologia
20.
Colloids Surf B Biointerfaces ; 236: 113794, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38382224

RESUMO

Targeting specific tumour cells and their microenvironments is essential for enhancing the efficacy of chemotherapy and reducing its side effects. A partial epithelial-to-mesenchymal transition state (pEMT, with a hybrid epithelial/mesenchymal phenotype) in tumour cells is an attractive targeting for anticancer treatment because it potentially provides maximal stemness and metastasis relevant to malignant cancer stem cell-like features. However, treatment strategies to target pEMT in tumour cells remain a challenge. This study demonstrates that extracellular cerium oxide nanoparticles (CNPs) selectively inhibit the growth of pEMT-induced tumour cells, without affecting full epithelial tumour cells. Herein, highly concentrated Ce3+ and Ce4+ ions are formed on CNP-layered poly-L-lactic acid surfaces. Cell cultures of pEMT-induced and uninduced lung cancer cell lines on the CNP-layered substrates allow the effect of extracellular CNPs on tumour cell growth to be investigated. The extracellular CNPs with dominant Ce3+ and Ce4+ ions were able to trap pEMT-induced tumour cells in a growth-arrested quiescent/dormant or cytostatic state without generating redox-related reactive oxygen species (ROS), i.e. non-redox mechanisms. The dominant Ce3+ state provided highly efficient growth inhibition of the pEMT-induced tumour cells. In contrast, the dominant Ce4+ state showed highly selective and appropriate growth regulation of normal and tumour cells, including a mesenchymal phenotype. Furthermore, Ce4+-CNPs readily adsorbed serum-derived fibronectin and laminin. Cerium valence-specific proteins adsorbed on CNPs may influence receptor-mediated cell-CNP interactions, leading to tumour cell growth inhibition. These findings provide new perspectives for pEMT-targeting anticancer treatments based on the unique biointerface of extracellular CNPs with different Ce valence states.


Assuntos
Cério , Nanopartículas , Oxirredução , Linhagem Celular , Cério/farmacologia , Íons
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA