Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Cell Sci ; 135(10)2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35510498

RESUMO

Distant metastasis mainly occurs through hematogenous dissemination, where suspended circulating tumor cells (CTCs) experience a considerable level of fluid shear stress. We recently reported that shear flow induced substantial apoptosis of CTCs, although a small subpopulation could still persist. However, how suspended tumor cells survive in shear flow remains poorly understood. This study finds that fluid shear stress eliminates the majority of suspended CTCs and increases nuclear size, whereas it has no effect on the viability of adherent tumor cells and decreases their nuclear size. Shear flow promotes histone acetylation in suspended tumor cells, the inhibition of which using one drug suppresses shear-induced nuclear expansion, suggesting that shear stress might increase nuclear size through histone acetylation. Suppressing histone acetylation-mediated nuclear expansion enhances shear-induced apoptosis of CTCs. These findings suggest that suspended tumor cells respond to shear stress through histone acetylation-mediated nuclear expansion, which protects CTCs from shear-induced destruction. Our study elucidates a unique mechanism underlying the mechanotransduction of suspended CTCs to shear flow, which might hold therapeutic promise for CTC eradication.


Assuntos
Células Neoplásicas Circulantes , Contagem de Células , Histonas , Humanos , Mecanotransdução Celular , Células Neoplásicas Circulantes/patologia , Estresse Mecânico
2.
Int J Mol Sci ; 24(8)2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-37108248

RESUMO

Tumor cells metastasize from a primary lesion to distant organs mainly through hematogenous dissemination, in which tumor cell re-adhesion to the endothelium is essential before extravasating into the target site. We thus hypothesize that tumor cells with the ability to adhere to the endothelium of a specific organ exhibit enhanced metastatic tropism to this target organ. This study tested this hypothesis and developed an in vitro model to mimic the adhesion between tumor cells and brain endothelium under fluid shear stress, which selected a subpopulation of tumor cells with enhanced adhesion strength. The selected cells up-regulated the genes related to brain metastasis and exhibited an enhanced ability to transmigrate through the blood-brain barrier. In the soft microenvironments that mimicked brain tissue, these cells had elevated adhesion and survival ability. Further, tumor cells selected by brain endothelium adhesion expressed higher levels of MUC1, VCAM1, and VLA-4, which were relevant to breast cancer brain metastasis. In summary, this study provides the first piece of evidence to support that the adhesion of circulating tumor cells to the brain endothelium selects the cells with enhanced brain metastasis potential.


Assuntos
Neoplasias Encefálicas , Neoplasias da Mama , Células Neoplásicas Circulantes , Humanos , Feminino , Neoplasias da Mama/metabolismo , Células Neoplásicas Circulantes/patologia , Endotélio/metabolismo , Adesão Celular , Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Metástase Neoplásica/patologia , Endotélio Vascular/metabolismo , Microambiente Tumoral
3.
Nano Lett ; 21(7): 3225-3236, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33764789

RESUMO

Developing strategies for efficient expansion of cancer stem-like cells (CSCs) in vitro will help investigate the mechanism underlying tumorigenesis and cancer recurrence. Herein, we report a dynamic culture substrate tethered with integrin ligand-bearing magnetic nanoparticles via a flexible polymeric linker to enable magnetic manipulation of the nanoscale ligand tether mobility. The cancer cells cultured on the substrate with high ligand tether mobility develop into large semispherical colonies with CSCs features, which can be abrogated by magnetically restricting the ligand tether mobility. Mechanistically, the substrate with high ligand tether mobility suppresses integrin-mediated mechanotransduction and histone-related methylation, thereby enhancing cancer cell stemness. The culture-derived high-stemness cells can generate tumors both locally and at the distant lung and uterus much more efficiently than the low-stemness cells. We believe that this magnetic nanoplatform provides a promising strategy for investigating the dynamic interaction between CSCs and the microenvironment and establishing a cost-effective tumor spheroid model.


Assuntos
Mecanotransdução Celular , Neoplasias , Linhagem Celular Tumoral , Feminino , Humanos , Integrinas , Ligantes , Células-Tronco Neoplásicas , Microambiente Tumoral
4.
Biophys J ; 120(16): 3498-3507, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34022236

RESUMO

Myopia is a common ocular disorder with significant alterations in the anterior ocular structure, including the cornea. The cell biophysical phenotype has been proposed to reflect the state of various diseases. However, the biophysical properties of corneal cells have not been characterized during myopia progression and their relationship with myopia remains unknown. This study characterizes the biophysical properties of corneal cells in normal, myopic, and recovered conditions, using two classical myopia models. Surprisingly, myopic corneal cells considerably reduce F-actin and microtubule content and cellular stiffness and generate elevated traction force compared with control cells. When myopia is restored to the healthy state, these biophysical properties are partially or fully restored to the levels of control cells. Furthermore, the level of chromatin condensation is significantly increased in the nucleus of myopic corneal cells and reduced to a level similar to healthy cells after recovery. These findings demonstrate that the reversible biophysical alterations of corneal cells reflect myopia progression, facilitating the study of the role of corneal cell biophysics in myopia.


Assuntos
Miopia , Biofísica , Córnea , Progressão da Doença , Humanos , Refração Ocular
6.
Analyst ; 145(3): 797-804, 2020 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-31894762

RESUMO

Detecting trace amounts of copper ions (Cu2+) is of high importance since copper is an essential element in the environment and the human body. Despite the recent advances in Cu2+ detection, the current approaches still suffer from insensitivity and lack of in situ detection in living cells. In the present work, a fluorescent nanosensor based on porphyrinic metal-organic framework nanoparticles (MOF-525 NPs) is proposed for sensitive and selective monitoring of Cu2+ in aqueous solution and living cells. The MOF-525 NPs with attractive properties, including ultrasmall size, good water dispersity and intense red fluorescence, are prepared via a facile and environment-friendly hydrothermal route. The fluorescence signal of MOF-525 NPs could be quenched statically by Cu2+ with high selectivity due to the strong affinity of Cu2+ to the porphyrin ligand in MOF-525. The proposed fluorescent nanosensor has a linear response in the range of 1.0-250 nM with a low detection limit of 220 pM. Furthermore, it is successfully employed for the detection of Cu2+ in water samples and the intracellular imaging of Cu2+ in living cells, demonstrating its great potential in the sensing and biological fields.


Assuntos
Cobre/análise , Estruturas Metalorgânicas/química , Microscopia de Fluorescência , Nanopartículas/química , Espectrometria de Fluorescência , Sobrevivência Celular/efeitos dos fármacos , Corantes Fluorescentes/química , Células HeLa , Humanos , Íons/química , Limite de Detecção , Nanopartículas/toxicidade , Água/química
7.
Int J Mol Sci ; 21(21)2020 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-33143160

RESUMO

Tumor cells metastasize to distal organs mainly through hematogenous dissemination, where they experience considerable levels of fluid shear stress. Epithelial-mesenchymal transition (EMT) plays a critical role in tumor metastasis. However, how fluid shear stress influences the EMT phenotype of circulating tumor cells (CTCs) in suspension has not been fully understood. The role of shear-induced EMT in cell survival under blood shear flow remains unclear. This study shows that the majority of breast CTCs underwent apoptosis under shear flow and the surviving cells exhibited mesenchymal phenotype, suggesting that fluid shear stress induces EMT. Mechanistically, fluid shear stress-activated Jun N-terminal kinase (JNK) signaling, inhibition/activation of which suppressed/promoted the EMT phenotype. In particular, shear flow facilitated the JNK-dependent transition of epithelial CTCs into the mesenchymal status and maintained the pre-existing mesenchymal cells. Importantly, the induction of EMT suppressed the pro-apoptosis gene p53 upregulated modulator of apoptosis (PUMA) and enhanced the survival of suspended CTCs in fluid shear stress, which was rescued by overexpressing PUMA or silencing JNK signaling, suggesting that shear-induced EMT promotes CTC survival through PUMA downregulation and JNK activation. Further, the expressions of EMT markers and JUN were correlated with poor patient survival. In summary, our findings have demonstrated that fluid shear stress induces EMT in suspended CTCs via JNK signaling that promotes their survival in shear flow. This study thus unveils a new role of blood shear stress in CTC survival and facilitates the development of novel therapeutics against tumor metastasis.


Assuntos
Fibroblastos Associados a Câncer/patologia , Neoplasias Colorretais/tratamento farmacológico , Transição Epitelial-Mesenquimal , Sistema de Sinalização das MAP Quinases , Células Neoplásicas Circulantes/patologia , Estresse Mecânico , Animais , Apoptose , Fibroblastos Associados a Câncer/metabolismo , Movimento Celular , Proliferação de Células , Colágeno/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Células Neoplásicas Circulantes/metabolismo , Fenótipo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biophys J ; 116(10): 1803-1814, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31076101

RESUMO

Tumor cells disseminate to distant organs mainly through blood circulation in which they experience considerable levels of fluid shear stress. However, the effects of hemodynamic shear stress on biophysical properties and functions of circulating tumor cells (CTCs) in suspension are not fully understood. In this study, we found that the majority of suspended breast tumor cells could be eliminated by fluid shear stress, whereas cancer stem cells held survival advantages over conventional cancer cells. Compared to untreated cells, tumor cells surviving shear stress exhibited unique biophysical properties: 1) cell adhesion was significantly retarded, 2) these cells exhibited elongated morphology and enhanced spreading and expressed genes related to epithelial-mesenchymal transition or hybrid phenotype, and 3) surviving tumor cells showed reduced F-actin assembly and stiffness. Importantly, inhibiting actomyosin activity promoted the survival of suspended tumor cells in fluid shear stress, whereas activating actomyosin suppressed cell survival, which might be explained by the up- and downregulation of the antiapoptosis genes. Soft surviving tumor cells held survival advantages in shear flow and higher resistance to chemotherapy. Inhibiting actomyosin activity in untreated cells enhanced chemoresistance, whereas activating actomyosin in surviving tumor cells suppressed this ability. These findings might be associated with the corresponding changes in the genes related to multidrug resistance. In summary, these data demonstrate that hemodynamic shear stress significantly influences biophysical properties and functions of suspended tumor cells. Our study unveils the regulatory roles of actomyosin in the survival and drug resistance of suspended tumor cells in hemodynamic shear flow, which suggest the importance of fluid shear stress and actomyosin activity in tumor metastasis. These findings may reveal a new, to our knowledge, mechanism by which CTCs are able to survive hemodynamic shear stress and chemotherapy and may offer a new potential strategy to target CTCs in shear flow and combat chemoresistance through actomyosin.


Assuntos
Actomiosina/metabolismo , Resistencia a Medicamentos Antineoplásicos , Resistência ao Cisalhamento , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Sobrevivência Celular , Hemodinâmica , Humanos , Estresse Mecânico
9.
Soft Matter ; 14(47): 9528-9533, 2018 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-30468439

RESUMO

Tumor cells disseminate to distant organs mainly through blood circulation, where they experience considerable levels of fluid shear flow. However, its influence on circulating tumor cells remains less understood. This study elucidates the effects of hemodynamic shear flow on biophysical properties and functions of breast circulating tumor cells with metastatic preference to brain. Only a small subpopulation of tumor cells are able to survive in shear flow with enhanced anti-apoptosis ability. Compared to untreated cells, surviving tumor cells spread more on soft substrates that mimic brain tissue but less on stiff substrates. They exhibit much lower expression of F-actin and cell stiffness but generate significantly higher cellular contractility. In addition, hemodynamic shear flow upregulates the stemness genes and considerably changes the expression of the genes related to brain metastasis. The enhanced cell spreading on soft substrates, reduced stiffness, elevated cellular contractility, and upregulation of the stemness and brain metastasis genes in tumor cells after shear flow treatment may be related to breast cancer metastasis in soft brain tissues. Our findings thus provide the first piece of evidence that hemodynamic shear flow regulates biophysical properties and functions of circulating tumor cells that are associated with brain metastasis, suggesting that tumor cells surviving in blood shear flow may better reflect the characteristics of organ preference in metastasis.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/secundário , Células Neoplásicas Circulantes/patologia , Feminino , Hemodinâmica/genética , Hemodinâmica/fisiologia , Humanos , Metástase Neoplásica/patologia , Estresse Mecânico
10.
Biochem Biophys Res Commun ; 483(1): 456-462, 2017 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-28007596

RESUMO

Tumor-repopulating cells (TRCs) are a tumorigenic sub-population of cancer cells that drives tumorigenesis. We have recently reported that soft fibrin matrices maintain TRC growth by promoting histone 3 lysine 9 (H3K9) demethylation and Sox2 expression and that Cdc42 expression influences H3K9 methylation. However, the underlying mechanisms of how soft matrices induce H3K9 demethylation remain elusive. Here we find that TRCs exhibit lower focal adhesion kinase (FAK) and H3K9 methylation levels in soft fibrin matrices than control melanoma cells on 2D rigid substrates. Silencing FAK in control melanoma cells decreases H3K9 methylation, whereas overexpressing FAK in tumor-repopulating cells enhances H3K9 methylation. Overexpressing Cdc42 or RhoA in the presence of FAK knockdown restores H3K9 methylation levels. Importantly, silencing FAK, Cdc42, or RhoA promotes Sox2 expression and proliferation of control melanoma cells in stiff fibrin matrices, whereas overexpressing each gene suppresses Sox2 expression and reduces growth of TRCs in soft but not in stiff fibrin matrices. Our findings suggest that low FAK mediated by soft fibrin matrices downregulates H3K9 methylation through reduction of Cdc42 and RhoA and promotes TRC growth.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Quinase 1 de Adesão Focal/genética , Histonas , Humanos , Lisina/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Metilação , Camundongos , Fatores de Transcrição SOXB1/metabolismo , Ensaio Tumoral de Célula-Tronco/métodos , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
12.
Acta Biomater ; 176: 321-333, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38272199

RESUMO

Hepatocellular carcinoma (HCC) cells, especially those with metastatic competence, show reduced stiffness compared to the non-malignant counterparts. However, it is still unclear whether and how the mechanics of HCC cells influence their migration and invasion. This study reports that HCC cells with enhanced motility show reduced mechanical stiffness and cytoskeleton, suggesting the inverse correlation between cellular stiffness and motility. Through pharmacologic and genetic approaches, inhibiting actomyosin activity reduces HCC cellular stiffness but promotes their migration and invasion, while activating it increases cell stiffness but impairs cell motility. Actomyosin regulates cell motility through the influence on cellular stiffness. Mechanistically, weakening/strengthening cells inhibits/promotes c-Jun N terminal kinase (JNK) phosphorylation, activation/inhibition of which rescues the effects of cell mechanics on their migration and invasion. Further, HCC cancer stem cells (CSCs) exhibit higher motility but lower stiffness than control cells. Increasing CSC stiffness weakens migration and invasion through the activation of JNK signaling. In conclusion, our findings unveil a new regulatory role of actomyosin-mediated cellular mechanics in tumor cell motility and present new evidence to support that tumor cell softening may be one driving force for HCC metastasis. STATEMENT OF SIGNIFICANCE: Tumor cells progressively become softened during metastasis and low cell stiffness is associated with high metastatic potential. However, it remains unclear whether tumor cell softening is a by-product of or a driving force for tumor progression. This work reports that the stiffness of hepatocellular carcinoma cells is linked to their migration and invasion. Importantly, tumor cell softening promotes migration and invasion, while cell stiffening impairs the mobility. Weakening/strengthening cells inhibits/promotes JNK phosphorylation, activation/inhibition of which rescues the effects of cell mechanics on their migration and invasion ability. Further, stiffening liver cancer stem cells attenuates their motility through activating JNK signaling. In summary, our study uncovers a previously unappreciated role of tumor cell mechanics in migration and invasion and implicates the therapeutic potential of cell mechanics in the mechanotargeting of metastasis.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Actomiosina , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Invasividade Neoplásica
13.
ACS Appl Mater Interfaces ; 16(7): 8289-8300, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38329794

RESUMO

ß-Tricalcium phosphate (ß-TCP) is a bioactive material for bone regeneration, but its brittleness limits its use as a standalone scaffold. Therefore, continuous efforts are necessary to effectively integrate ß-TCP into polymers, facilitating a sturdy ion exchange for cell regulation. Herein, a novel semiembedded technique was utilized to anchor ß-TCP nanoparticles onto the surface of the elastic polymer, followed by hydrophilic modification with the polymerization of dopamine. Cell adhesion and osteogenic differentiation of mesenchymal stem cells (MSCs) under static and dynamic uniaxial cyclic stretching conditions were investigated. The results showed that the new strategy was effective in promoting cell adhesion, proliferation, and osteogenic induction by the sustained release of Ca2+ in the vicinity and creating a reasonable roughness. Specifically, released Ca2+ from ß-TCP could activate the calcium signaling pathway, which further upregulated calmodulin and calcium/calmodulin-dependent protein kinase II genes in MSCs. Meanwhile, the roughness of the membrane and the uniaxial cyclic stretching activated the PIEZO1 signaling pathway. Chemical and mechanical stimulation promotes osteogenic differentiation and increases the expression of related genes 2-8-fold. These findings demonstrated that the neoteric semiembedded structure was a promising strategy in controlling both chemical and mechanical factors of biomaterials for cell regulation.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Fosfatos de Cálcio/química , Diferenciação Celular , Alicerces Teciduais
14.
Cancer Lett ; 590: 216870, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38614386

RESUMO

To seed lethal secondary lesions, circulating tumor cells (CTCs) must survive all rate-limiting factors during hematogenous dissemination, including fluid shear stress (FSS) that poses a grand challenge to their survival. We thus hypothesized that CTCs with the ability to survive FSS in vasculature might hold metastasis-initiating competence. This study reported that FSS of physiologic magnitude selected a small subpopulation of suspended tumor cells in vitro with the traits of metastasis-initiating cells, including stemness, migration/invasion potential, cellular plasticity, and biophysical properties. These shear-selected cells generated local and metastatic tumors at the primary and distal sites efficiently, implicating their metastasis competence. Mechanistically, FSS activated the mechanosensitive protein CXCR4 and the downstream PI3K/AKT signaling, which were essential in shear-mediated selection of metastasis-competent CTCs. In summary, these findings conclude that CTCs with metastasis-initiating competence survive FSS during hematogenous dissemination through CXCR4-PI3K/AKT signaling, which may provide new therapeutic targets for the early prevention of tumor metastasis.


Assuntos
Células Neoplásicas Circulantes , Transdução de Sinais , Animais , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Movimento Celular , Metástase Neoplásica , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores CXCR4/metabolismo , Estresse Mecânico
15.
Nat Mater ; 11(8): 734-41, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22751180

RESUMO

The identification of stem-cell-like cancer cells through conventional methods that depend on stem cell markers is often unreliable. We developed a mechanical method for selecting tumorigenic cells by culturing single cancer cells in fibrin matrices of ~100 Pa in stiffness. When cultured within these gels, primary human cancer cells or single cancer cells from mouse or human cancer cell lines grew within a few days into individual round colonies that resembled embryonic stem cell colonies. Subcutaneous or intravenous injection of 10 or 100 fibrin-cultured cells in syngeneic or severe combined immunodeficiency mice led to the formation of solid tumours at the site of injection or at the distant lung organ much more efficiently than control cancer cells selected using conventional surface marker methods or cultured on conventional rigid dishes or on soft gels. Remarkably, as few as ten such cells were able to survive and form tumours in the lungs of wild-type non-syngeneic mice.


Assuntos
Separação Celular/métodos , Fibrina , Géis , Melanoma Experimental/patologia , Células-Tronco Neoplásicas , Esferoides Celulares , Animais , Sobrevivência Celular , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos SCID , Células Tumorais Cultivadas
16.
APL Bioeng ; 7(3): 036108, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37575881

RESUMO

Tumor cells metastasize to distant organs mainly via hematogenous dissemination, in which circulating tumor cells (CTCs) are relatively vulnerable, and eliminating these cells has great potential to prevent metastasis. In vasculature, natural killer (NK) cells are the major effector lymphocytes for efficient killing of CTCs under fluid shear stress (FSS), which is an important mechanical cue in tumor metastasis. However, the influence of FSS on the cytotoxicity of NK cells against CTCs remains elusive. We report that the death rate of CTCs under both NK cells and FSS is much higher than the combined death induced by either NK cells or FSS, suggesting that FSS may enhance NK cell's cytotoxicity. This death increment is elicited by shear-induced NK activation and granzyme B entry into target cells rather than the death ligand TRAIL or secreted cytokines TNF-α and IFN-γ. When NK cells form conjugates with CTCs or adhere to MICA-coated substrates, NK cell activating receptor NKG2D can directly sense FSS to induce NK activation and degranulation. These findings reveal the promotive effect of FSS on NK cell's cytotoxicity toward CTCs, thus providing new insight into immune surveillance of CTCs within circulation.

17.
Oncogene ; 42(47): 3457-3490, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37864030

RESUMO

Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.


Assuntos
Mecanotransdução Celular , Neoplasias , Humanos , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais , Microambiente Tumoral , Biofísica
18.
Sci Adv ; 9(27): eadg9593, 2023 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-37418519

RESUMO

Biophysical cues of rigid tumor matrix play a critical role in cancer cell malignancy. We report that stiffly confined cancer cells exhibit robust growth of spheroids in the stiff hydrogel that exerts substantial confining stress on the cells. The stressed condition activated Hsp (heat shock protein)-signal transducer and activator of transcription 3 signaling via the transient receptor potential vanilloid 4-phosphatidylinositol 3-kinase/Akt axis, thereby up-regulating the expression of the stemness-related markers in cancer cells, whereas these signaling activities were suppressed in cancer cells cultured in softer hydrogels or stiff hydrogels with stress relief or Hsp70 knockdown/inhibition. This mechanopriming based on three-dimensional culture enhanced cancer cell tumorigenicity and metastasis in animal models upon transplantation, and pharmaceutically inhibiting Hsp70 improved the anticancer efficacy of chemotherapy. Mechanistically, our study reveals the crucial role of Hsp70 in regulating cancer cell malignancy under mechanically stressed conditions and its impacts on cancer prognosis-related molecular pathways for cancer treatments.


Assuntos
Proteínas de Choque Térmico , Neoplasias , Animais , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Transdução de Sinais , Proteínas de Choque Térmico HSP70/metabolismo , Hidrogéis , Linhagem Celular Tumoral
19.
Research (Wash D C) ; 6: 0224, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37746658

RESUMO

Tumor cells progressively remodel cytoskeletal structures and reduce cellular stiffness during tumor progression, implicating the correlation between cell mechanics and malignancy. However, the roles of tumor cell cytoskeleton and the mechanics in tumor progression remain incompletely understood. We report that softening/stiffening tumor cells by targeting actomyosin promotes/suppresses self-renewal in vitro and tumorigenic potential in vivo. Weakening/strengthening actin cytoskeleton impairs/reinforces the interaction between adenomatous polyposis coli (APC) and ß-catenin, which facilitates ß-catenin nuclear/cytoplasmic localization. Nuclear ß-catenin binds to the promoter of Oct4, which enhances its transcription that is crucial in sustaining self-renewal and malignancy. These results demonstrate that the mechanics of tumor cells dictate self-renewal through cytoskeleton-APC-Wnt/ß-catenin-Oct4 signaling, which are correlated with tumor differentiation and patient survival. This study unveils an uncovered regulatory role of cell mechanics in self-renewal and malignancy, and identifies tumor cell mechanics as a hallmark not only for cancer diagnosis but also for mechanotargeting.

20.
Front Bioeng Biotechnol ; 10: 953590, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263359

RESUMO

The cornea is a transparent, dome-shaped structure on the front part of the eye that serves as a major optic element and a protector from the external environment. Recent evidence shows aberrant alterations of the corneal mechano-environment in development and progression of various corneal diseases. It is, thus, critical to understand how corneal cells sense and respond to mechanical signals in physiological and pathological conditions. In this review, we summarize the corneal mechano-environment and discuss the impact of these mechanical cues on cellular functions from the bench side (in a laboratory research setting). From a clinical perspective, we comprehensively review the mechanical changes of corneal tissue in several cornea-related diseases, including keratoconus, myopia, and keratectasia, following refractive surgery. The findings from the bench side and clinic underscore the involvement of mechanical cues in corneal disorders, which may open a new avenue for development of novel therapeutic strategies by targeting corneal mechanics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA