1
|
Lee YL, Longmore GD, Pathak A. Distinct roles of protrusions and collagen deformation in collective invasion of cancer cell types. Biophys J 2025; 124:1506-1520. [PMID: 40170350 DOI: 10.1016/j.bpj.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
The breast tumor microenvironment is composed of heterogeneous cell populations, including normal epithelial cells, cancer-associated fibroblasts (CAFs), and tumor cells that lead collective cell invasion. Both leader tumor cells and CAFs are known to play important roles in tumor invasion across the collagen-rich stromal boundary. However, their individual abilities to utilize their cell-intrinsic protrusions and perform force-based collagen remodeling to collectively invade remain unclear. To compare collective invasion phenotypes of leader-like tumor cells and CAFs, we embedded spheroids composed of 4T1 tumor cells or mouse tumor-derived CAF cell lines within 3D collagen gels and analyzed their invasion and collagen deformation. We found that 4T1s undergo greater invasion while generating lower collagen deformation compared with CAFs. Although force-driven collagen deformations are conventionally associated with higher cellular forces and invasion, here 4T1s specifically rely on actin-based protrusions, while CAFs rely on myosin-based contractility for collective invasion. In denser collagen, both cell types slowed their invasion, and selective pharmacological inhibitions show that Arp2/3 is required but myosin-II is dispensable for 4T1 invasion. Furthermore, depletion of CDH3 from 4T1s and DDR2 from CAFs reduces their ability to distinguish between collagen densities. For effective invasion, both cell types reorient and redistribute magnetically prealigned collagen fibers. With heterogeneous cell populations of cocultured CAFs and 4T1s, higher percentage of CAFs impeded invasion while increasing collagen fiber alignment. Overall, our findings demonstrate distinctive mechanisms of collective invasion adopted by 4T1 tumor cells and CAFs, one relying more on protrusions and the other on force-based collagen deformation. These results suggest that individually targeting cellular protrusions or contractility may not be universally applicable for all cell types or collagen densities, and a better cell-type-dependent approach could enhance effectiveness of cancer therapies.
Collapse
Affiliation(s)
- Ye Lim Lee
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Gregory D Longmore
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri; ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri.
| |
Collapse
|
2
|
Canário R, Ribeiro AS, Morgado I, Peixoto A, Barbosa A, Santos C, Mendes N, Lopes P, Monteiro P, Coelho R, Jacob F, Heinzelmann-Schwarz V, Ricardo S, Teixeira MR, Bartosch C, Paredes J. P-cadherin overexpression is associated with early transformation of the Fallopian tube epithelium and aggressiveness of tubo-ovarian high-grade serous carcinoma. Virchows Arch 2025:10.1007/s00428-025-04104-7. [PMID: 40320493 DOI: 10.1007/s00428-025-04104-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 05/20/2025]
Abstract
Tubo-ovarian high-grade serous carcinoma (HGSC) with proficient homologous recombination (HR) DNA repair (HRP) accounts for approximately 50% of cases and is associated with platinum-resistance and poor prognosis. We hypothesize that the acquisition of hybrid phenotypes displaying both epithelial and mesenchymal (E/M) features may be involved in the malignant transformation and tumour dissemination in this subgroup. Therefore, we analysed, by digital pathology, the expression and prognostic significance of 3 classic cadherins (E-cadherin, epithelial marker; N-cadherin, mesenchymal marker; and P-cadherin, candidate marker of hybrid E/M) in 577 formalin-fixed paraffin-embedded human samples representing the putative stepwise serous carcinogenesis in the Fallopian tube epithelium (FTE). We observed a non-canonical N-to-P-cadherin switch along the carcinogenic progression, with a statistically significant overexpression of P-cadherin in pre-malignant and malignant samples, compared to the control FTE. Interestingly, this overexpression was most pronounced in precursor lesions and HGSC cells from malignant ascites. Tumours with high P-cadherin expression were significantly associated with worse overall survival, especially in the subgroup without BRCA1/2 mutations. Transient P-cadherin knock-down resulted in in vitro significant reduction of functional hybrid E/M hallmarks, namely decreased anoikis resistance, reduced collective migration and invasion in a representative platinum-resistant HRP cell line. Taken together, our results suggest that P-cadherin overexpression is an early event in the serous carcinogenesis and may be involved in hybrid E/M activation in HRP-HGSC, further supporting this adhesion molecule as a promising biomarker for this poor prognostic subgroup.
Collapse
Affiliation(s)
- Rita Canário
- Graduate Program in Areas of Basic and Applied Biology (GABBA), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Cancer Metastasis, i3S-Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Cancer Genetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
| | - Ana Sofia Ribeiro
- Cancer Metastasis, i3S-Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Inês Morgado
- Cancer Metastasis, i3S-Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Ana Peixoto
- Cancer Genetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
| | - Ana Barbosa
- Cancer Genetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
| | - Catarina Santos
- Cancer Genetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
| | - Nuno Mendes
- Histology and Electron Microscopy, i3S-Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Paula Lopes
- Cancer Biology and Epigenetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca) , Porto, Portugal
| | - Paula Monteiro
- Cancer Biology and Epigenetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca) , Porto, Portugal
| | - Ricardo Coelho
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031, Basel, Switzerland
| | - Francis Jacob
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031, Basel, Switzerland
| | - Viola Heinzelmann-Schwarz
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031, Basel, Switzerland
| | - Sara Ricardo
- Ageing and Stress Group, i3S-Institute for Research and Innovation in Health,, University of Porto, Porto, Portugal
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences-CESPU, Gandra, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, Portugal
| | - Manuel R Teixeira
- Cancer Genetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Pathology and Molecular Immunology Department, School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Carla Bartosch
- Cancer Biology and Epigenetics Group, Research Center-Portuguese Oncology Institute of Porto (CI-IPO-Porto)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca), Porto, Portugal
- Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto. Comprehensive Cancer Center Raquel Seruca (P.CCC Raquel Seruca) , Porto, Portugal
- Pathology and Molecular Immunology Department, School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Joana Paredes
- Cancer Metastasis, i3S-Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.
- FMUP-Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
3
|
Tran SK, Lichtenberg JY, Leonard CE, Williamson JR, Sterling HR, Panek GK, Pearson AH, Lopez S, Lemmon CA, Conway DE, Hwang PY. P-cadherin-dependent adhesions are required for single lumen formation and HGF-mediated cell protrusions during epithelial morphogenesis. iScience 2025; 28:111844. [PMID: 39981519 PMCID: PMC11840494 DOI: 10.1016/j.isci.2025.111844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/09/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
During epithelial morphogenesis, in vivo, epithelial cells form cysts enclosing a single, hollow lumen and extend protrusions as a precursor for tubulogenesis. Cell-cell adhesions (e.g., cadherins) contribute to successful execution of these processes; while there are many different cadherins, one less studied cadherin in epithelial morphogenesis is P-cadherin (CDH3). Here, we investigated the role of CDH3 on successful lumen formation and cell protrusions, using three-dimensional cultures of Madin-Darby canine kidney (MDCK) and CDH3 knockout cell lines. We show that depletion of CDH3 leads to perturbations of hollow lumen formation associated with defects in cell protrusions and tubulogenesis, mediated by Rho/ROCK pathway. CDH3 knockout cells exert lower forces on the surrounding environment compared to wild-type cells, suggesting CDH3 acts as a mechanosensor for stable cell protrusion establishment. Together, our data suggest that CDH3 has an essential function during epithelial morphogenesis by contributing to lumen formation and cell protrusions.
Collapse
Affiliation(s)
- Sydnie K. Tran
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Jessanne Y. Lichtenberg
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Corinne E. Leonard
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Jessica R. Williamson
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Hazel R. Sterling
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Grace K. Panek
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA
| | - Amanda H. Pearson
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Santiago Lopez
- Department of Biomedical Engineering, Rice University, Houston, TX 77251, USA
| | - Christopher A. Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Daniel E. Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Priscilla Y. Hwang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
4
|
Leng D, Yang M, Miao X, Huang Z, Li M, Liu J, Wang T, Li D, Feng C. Dynamic changes in the skin transcriptome for the melanin pigmentation in embryonic chickens. Poult Sci 2025; 104:104210. [PMID: 39693959 PMCID: PMC11720608 DOI: 10.1016/j.psj.2024.104210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/12/2024] [Accepted: 08/08/2024] [Indexed: 12/20/2024] Open
Abstract
Dermal hyperpigmentation stands out among the various skin pigmentation phenotypes in chickens, where most other pigmentation variants affect feather color and patterning predominantly. Despite numerous black chicken breeds worldwide, only a select few exhibit comprehensive black pigmentation, which encompasses the skin, meat, flesh, and bones. The process of skin melanin pigmentation is intricate and develops successively. Historically, research has concentrated primarily on specific developmental points or stages, but fewer studies have examined the entire transcriptome across the timeline of the development of the embryo integument. In our investigation, we undertook the sequencing of chicken embryo skin samples from d 4 to d 13 of incubation. Our results showed that melanoblasts continued to migrate from E4 to the epidermis until E12. Beginning with E6, melanin was synthesized and transferred to epidermal cells and feather follicles in large quantities, and genes such as DCT, TYR, TYRP1, and MITF played a key role in this process, which is significantly different from that of white-skinned chickens. There were 854 differentially expressed genes between E7 and E8. At this stage, melanocytes formed dendritic forms and transferred melanin to keratinocytes, while the dorsal skin became visibly dark. In addition, CDH3, which is a core factor involved in a variety of biological processes, may have an important impact on skin melanin pigmentation. Collectively, our findings unveiled a phased relationship between the canonical pathway and the noncanonical pathway from E4 to E13. These analyses illuminated the gene regulatory mechanism and provided foundational data that pertained to pigmentation in chickens.
Collapse
Affiliation(s)
- Dong Leng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Maosen Yang
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Xiaomeng Miao
- Institute of Animal Husbandry and Veterinary Medicine, Guizhou Academy of Agricultural Sciences, Guiyang 550005, China
| | - Zhiying Huang
- School of Pharmacy, Chengdu University, Chengdu 610106, China; College of Animal Science, Shanxi Agricultural University, Taiyuan 030031, China
| | - Mengmeng Li
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jia Liu
- Guizhou Province Livestock and Poultry Genetic Resources Management Station, Guizhou Provincial Department of Agriculture and Rural Affairs, Guiyang 550001, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Chungang Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
5
|
Berdiaki A, Neagu M, Tzanakakis P, Spyridaki I, Pérez S, Nikitovic D. Extracellular Matrix Components and Mechanosensing Pathways in Health and Disease. Biomolecules 2024; 14:1186. [PMID: 39334952 PMCID: PMC11430160 DOI: 10.3390/biom14091186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Glycosaminoglycans (GAGs) and proteoglycans (PGs) are essential components of the extracellular matrix (ECM) with pivotal roles in cellular mechanosensing pathways. GAGs, such as heparan sulfate (HS) and chondroitin sulfate (CS), interact with various cell surface receptors, including integrins and receptor tyrosine kinases, to modulate cellular responses to mechanical stimuli. PGs, comprising a core protein with covalently attached GAG chains, serve as dynamic regulators of tissue mechanics and cell behavior, thereby playing a crucial role in maintaining tissue homeostasis. Dysregulation of GAG/PG-mediated mechanosensing pathways is implicated in numerous pathological conditions, including cancer and inflammation. Understanding the intricate mechanisms by which GAGs and PGs modulate cellular responses to mechanical forces holds promise for developing novel therapeutic strategies targeting mechanotransduction pathways in disease. This comprehensive overview underscores the importance of GAGs and PGs as key mediators of mechanosensing in maintaining tissue homeostasis and their potential as therapeutic targets for mitigating mechano-driven pathologies, focusing on cancer and inflammation.
Collapse
Affiliation(s)
- Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Monica Neagu
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
| | - Petros Tzanakakis
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Ioanna Spyridaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Serge Pérez
- Centre de Recherche sur les Macromolécules Végétales (CERMAV), Centre National de la Recherche Scientifique (CNRS), University Grenoble Alpes, 38000 Grenoble, France;
| | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| |
Collapse
|
6
|
Ma W, Hu J. Downregulated CDH3 is correlated with a better prognosis for LUAD and decreases proliferation and migration of lung cancer cells. Genes Genomics 2024; 46:713-731. [PMID: 38064156 DOI: 10.1007/s13258-023-01476-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 11/05/2023] [Indexed: 05/19/2024]
Abstract
BACKGROUND CDH3 is a glycoprotein with a single-span transmembrane domain that mediates cell-to-cell adhesion. Abnormal expression of CDH3 is associated with a poor prognosis in patients with breast, thyroid, colorectal carcinomas and glioblastoma. Soluble CDH3 in pleural effusions can be used as a marker for real-time monitoring of resistance to first- and second-generation EGFR-TKIs. The CDH3 mechanism underlying lung adenocarcinomas (LUADs) has not been established. OBJECTIVE This study analyzed the correlation between CDH3 expression and lung cancer prognosis and the effect of down-regulation CDH3 expression on the proliferation and migration of lung cancer cells. METHODS CDH3 expression was studied using the Oncomine, TIMER, PanglaoDB, and GEPIA databases. The effect of CDH3 on clinical prognosis was assessed with GEPIA, the PrognoScan database, and Kaplan-Meier plotter. The relationship between CDH3 to immune infiltrating cells was explored using TIMER and TISIDB. The function of CDH3 in lung cancer cell lines was determined by CCK-8 and wound healing assays in vitro. Furthermore, RNA sequencing was used to identify key signaling pathways and differentially-expressed genes. RESULTS LUAD tissues had higher CDH3 expression compared with normal tissues and were associated with worse overall survival in patients with LUAD. CDH3 expression had positive associations with infiltration of CD4 + T cells, Tregs and exhausted T cells, but negative associations with infiltration of B cells in patients with LUAD. CCK-8 and wound healing assays revealed that downregulation of CDH3 inhibited the proliferation and migration of cells. KEGG analysis revealed that the TGF-beta signaling pathways were demonstrated to be enriched pathways for genes negatively regulated by knockdown of CDH3. CONCLUSION CDH3 expression affects proliferation and migration of lung cancer cells and might serve as a potential prognostic marker in LUAD patients.
Collapse
Affiliation(s)
- Wanru Ma
- Department of Blood Transfusion, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Da Hua Road, Dong Dan, Beijing, 100730, People's Republic of China
| | - Junhua Hu
- Department of Blood Transfusion, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Da Hua Road, Dong Dan, Beijing, 100730, People's Republic of China.
| |
Collapse
|
7
|
Lee YL, Mathur J, Walter C, Zmuda H, Pathak A. Matrix obstructions cause multiscale disruption in collective epithelial migration by suppressing leader cell function. Mol Biol Cell 2023; 34:ar94. [PMID: 37379202 PMCID: PMC10398892 DOI: 10.1091/mbc.e22-06-0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/06/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
During disease and development, physical changes in extracellular matrix cause jamming, unjamming, and scattering in epithelial migration. However, whether disruptions in matrix topology alter collective cell migration speed and cell-cell coordination remains unclear. We microfabricated substrates with stumps of defined geometry, density, and orientation, which create obstructions for migrating epithelial cells. Here, we show that cells lose their speed and directionality when moving through densely spaced obstructions. Although leader cells are stiffer than follower cells on flat substrates, dense obstructions cause overall cell softening. Through a lattice-based model, we identify cellular protrusions, cell-cell adhesions, and leader-follower communication as key mechanisms for obstruction-sensitive collective cell migration. Our modeling predictions and experimental validations show that cells' obstruction sensitivity requires an optimal balance of cell-cell adhesions and protrusions. Both MDCK (more cohesive) and α-catenin-depleted MCF10A cells were less obstruction sensitive than wild-type MCF10A cells. Together, microscale softening, mesoscale disorder, and macroscale multicellular communication enable epithelial cell populations to sense topological obstructions encountered in challenging environments. Thus, obstruction-sensitivity could define "mechanotype" of cells that collectively migrate yet maintain intercellular communication.
Collapse
Affiliation(s)
- Ye Lim Lee
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Christopher Walter
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Hannah Zmuda
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
8
|
Zhu W, Qian W, Liao W, Huang X, Xu J, Qu W, Xue J, Feng F, Liu W, Liu F, Han L. Non-Invasive and Real-Time Monitoring of the Breast Cancer Metastasis Degree via Metabolomics. Cancers (Basel) 2022; 14:cancers14225589. [PMID: 36428687 PMCID: PMC9688400 DOI: 10.3390/cancers14225589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/19/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer (BC) is a serious threat to women's health and metastasis is the major cause of BC-associated mortality. Various techniques are currently used to preoperatively describe the metastatic status of tumors, based on which a comprehensive treatment protocol was determined. However, accurately staging a tumor before surgery remains a challenge, which may lead to the miss of optimal treatment options. More severely, the failure to detect and remove occult micrometastases often causes tumor recurrences. There is an urgent need to develop a more precise and non-invasive strategy for the detection of the tumor metastasis in lymph nodes and distant organs. Based on the facts that tumor metastasis is closely related to the primary tumor microenvironment (TME) evolutions and that metabolomics profiling of the circulatory system can precisely reflect subtle changes within TME, we suppose whether metabolomic technology can be used to achieve non-invasive and real-time monitoring of BC metastatic status. In this study, the metastasis status of BC mouse models with different tumor-bearing times was firstly depicted to mimic clinical anatomic TNM staging system. Metabolomic profiling together with metastasis-related changes in TME among tumor-bearing mice with different metastatic status was conducted. A range of differential metabolites reflecting tumor metastatic states were screened and in vivo experiments proved that two main metastasis-driving factors in TME, TGF-β and hypoxia, were closely related to the regular changes of these metabolites. The differential metabolites level changes were also preliminarily confirmed in a limited number of clinical BC samples. Metabolite lysoPC (16:0) was found to be useful for clinical N stage diagnosis and the possible cause of its changes was analyzed by bioinformatics techniques.
Collapse
Affiliation(s)
- Wanfang Zhu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Wenxin Qian
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Wenting Liao
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoxian Huang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Jiawen Xu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Jingwei Xue
- Tumor Precise Intervention and Translational Medicine Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an 271000, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
- School of Pharmacy, Nanjing Medical University, Nanjing 210029, China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
- Zhejiang Center for Safety Study of Drug Substances (Industrial Technology Innovation Platform), Hangzhou 310018, China
| | - Fulei Liu
- Tumor Precise Intervention and Translational Medicine Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an 271000, China
- Pharmaceutical Department, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an 271000, China
- Correspondence: (F.L.); (L.H.)
| | - Lingfei Han
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (F.L.); (L.H.)
| |
Collapse
|
9
|
Time-Series Clustering of Single-Cell Trajectories in Collective Cell Migration. Cancers (Basel) 2022; 14:cancers14194587. [PMID: 36230509 PMCID: PMC9559181 DOI: 10.3390/cancers14194587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/11/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary In this study, we normalized trajectories containing both mesenchymal and epithelial cells to remove the effect of cell location on clustering, and performed a dimensionality reduction on the time series data before clustering. When the clustering results were superimposed on the trajectories prior to normalization, the results still showed similarities in location, indicating that this method can find cells with similar migration patterns. These data highlight the reliability of this method in identifying consistent migration patterns in collective cell migration. Abstract Collective invasion drives multicellular cancer cells to spread to surrounding normal tissues. To fully comprehend metastasis, the methodology of analysis of individual cell migration in tissue should be well developed. Extracting and classifying cells with similar migratory characteristics in a colony would facilitate an understanding of complex cell migration patterns. Here, we used electrospun fibers as the extracellular matrix for the in vitro modeling of collective cell migration, clustering of mesenchymal and epithelial cells based on trajectories, and analysis of collective migration patterns based on trajectory similarity. We normalized the trajectories to eliminate the effect of cell location on clustering and used uniform manifold approximation and projection to perform dimensionality reduction on the time-series data before clustering. When the clustering results were superimposed on the trajectories before normalization, the results still exhibited positional similarity, thereby demonstrating that this method can identify cells with similar migration patterns. The same cluster contained both mesenchymal and epithelial cells, and this result was related to cell location and cell division. These data highlight the reliability of this method in identifying consistent migration patterns during collective cell migration. This provides new insights into the epithelial–mesenchymal interactions that affect migration patterns.
Collapse
|
10
|
P-Cadherin Regulates Intestinal Epithelial Cell Migration and Mucosal Repair, but Is Dispensable for Colitis Associated Colon Cancer. Cells 2022; 11:cells11091467. [PMID: 35563773 PMCID: PMC9100778 DOI: 10.3390/cells11091467] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 12/16/2022] Open
Abstract
Recurrent chronic mucosal inflammation, a characteristic of inflammatory bowel diseases (IBD), perturbs the intestinal epithelial homeostasis resulting in formation of mucosal wounds and, in most severe cases, leads to colitis-associated colon cancer (CAC). The altered structure of epithelial cell-cell adhesions is a hallmark of intestinal inflammation contributing to epithelial injury, repair, and tumorigenesis. P-cadherin is an important adhesion protein, poorly expressed in normal intestinal epithelial cells (IEC) but upregulated in inflamed and injured mucosa. The goal of this study was to investigate the roles of P-cadherin in regulating intestinal inflammation and CAC. P-cadherin expression was markedly induced in the colonic epithelium of human IBD patients and CAC tissues. The roles of P-cadherin were investigated in P-cadherin null mice using dextran sulfate sodium (DSS)-induced colitis and an azoxymethane (AOM)/DSS induced CAC. Although P-cadherin knockout did not affect the severity of acute DSS colitis, P-cadherin null mice exhibited faster recovery after colitis. No significant differences in the number of colonic tumors were observed in P-cadherin null and control mice. Consistently, the CRISPR/Cas9-mediated knockout of P-cadherin in human IEC accelerated epithelial wound healing without affecting cell proliferation. The accelerated migration of P-cadherin depleted IEC was driven by activation of Src kinases, Rac1 GTPase and myosin II motors and was accompanied by transcriptional reprogramming of the cells. Our findings highlight P-cadherin as a negative regulator of IEC motility in vitro and mucosal repair in vivo. In contrast, this protein is dispensable for IEC proliferation and CAC development.
Collapse
|
11
|
Bandzerewicz A, Gadomska-Gajadhur A. Into the Tissues: Extracellular Matrix and Its Artificial Substitutes: Cell Signalling Mechanisms. Cells 2022; 11:914. [PMID: 35269536 PMCID: PMC8909573 DOI: 10.3390/cells11050914] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
The existence of orderly structures, such as tissues and organs is made possible by cell adhesion, i.e., the process by which cells attach to neighbouring cells and a supporting substance in the form of the extracellular matrix. The extracellular matrix is a three-dimensional structure composed of collagens, elastin, and various proteoglycans and glycoproteins. It is a storehouse for multiple signalling factors. Cells are informed of their correct connection to the matrix via receptors. Tissue disruption often prevents the natural reconstitution of the matrix. The use of appropriate implants is then required. This review is a compilation of crucial information on the structural and functional features of the extracellular matrix and the complex mechanisms of cell-cell connectivity. The possibilities of regenerating damaged tissues using an artificial matrix substitute are described, detailing the host response to the implant. An important issue is the surface properties of such an implant and the possibilities of their modification.
Collapse
|
12
|
Di Martino JS, Akhter T, Bravo-Cordero JJ. Remodeling the ECM: Implications for Metastasis and Tumor Dormancy. Cancers (Basel) 2021; 13:4916. [PMID: 34638400 PMCID: PMC8507703 DOI: 10.3390/cancers13194916] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022] Open
Abstract
While most primary tumors can be effectively treated, therapeutics fail to efficiently eliminate metastases. Metastases arise from cancer cells that leave the primary tumor and seed distant sites. Recent studies have shown that cancer cells disseminate early during tumor progression and can remain dormant for years before they resume growth. In these metastatic organs, cancer cells reside in microenvironments where they interact with other cells, but also with the extracellular matrix (ECM). The ECM was long considered to be an inert, non-cellular component of tissues, providing their architecture. However, in recent years, a growing body of evidence has shown that the ECM is a key driver of cancer progression, and it can exert effects on tumor cells, regulating their metastatic fate. ECM remodeling and degradation is required for the early steps of the metastatic cascade: invasion, tumor intravasation, and extravasation. Similarly, ECM molecules have been shown to be important for metastatic outgrowth. However, the role of ECM molecules on tumor dormancy and their contribution to the dormancy-supportive niches is not well understood. In this perspective article, we will summarize the current knowledge of ECM and its role in tumor metastasis and dormancy. We will discuss how a better understanding of the individual components of the ECM niche and their roles mediating the dormant state of disseminated tumor cells (DTCs) will advance the development of new therapies to target dormant cells and prevent metastasis outgrowth.
Collapse
Affiliation(s)
| | | | - Jose Javier Bravo-Cordero
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.S.D.M.); (T.A.)
| |
Collapse
|
13
|
Peng Y, Meng G, Sheng X, Gao H. Transcriptome and DNA methylation analysis reveals molecular mechanisms underlying intrahepatic cholangiocarcinoma progression. J Cell Mol Med 2021; 25:6373-6387. [PMID: 34013637 PMCID: PMC8256365 DOI: 10.1111/jcmm.16615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is an aggressive malignancy with increasing incidence. It has been suggested that DNA methylation drives cancer development. However, the molecular mechanisms underlying iCCA progression and the roles of DNA methylation still remain elusive. In this study, weighted correlation networks were constructed to identify gene modules and hub genes associated with the tumour stage. We identified 12 gene modules, two of which were significantly positively or negatively related to the tumour stage, respectively. Key hub genes SLC2A1, CDH3 and EFHD2 showed increased expression across the tumour stage and were correlated with poor survival, whereas decrease of FAM171A1, ONECUT1 and PHYHIPL was correlated with better survival. Pathway analysis revealed hedgehog pathway was activated in CDH3 up-regulated tumours, and chromosome separation was elevated in tumours expressing high EFHD2. JAK-STAT pathway was overrepresented in ONECUT1 down-regulated tumours, whereas Rho GTPases-formins signalling was activated in PHYHIPL down-regulated tumours. Finally, significant negative associations between expression of EFHD2, PHYHIPL and promoter DNA methylation were detected, and alterations of DNA methylation were correlated with tumour survival. In summary, we identified key genes and pathways that may participate in progression of iCCA and proposed putative roles of DNA methylation in iCCA.
Collapse
Affiliation(s)
- Yuming Peng
- First Department of General SurgeryHunan Children’s HospitalChangshaChina
| | - Guohao Meng
- Department of PathophysiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiaotong University School of MedicineShanghaiChina
| | - Xinyi Sheng
- First Department of General SurgeryHunan Children’s HospitalChangshaChina
| | - Hongqiang Gao
- First Department of General SurgeryHunan Children’s HospitalChangshaChina
| |
Collapse
|
14
|
Clegg J, Koch MK, Thompson EW, Haupt LM, Kalita-de Croft P, Bray LJ. Three-Dimensional Models as a New Frontier for Studying the Role of Proteoglycans in the Normal and Malignant Breast Microenvironment. Front Cell Dev Biol 2020; 8:569454. [PMID: 33163489 PMCID: PMC7581852 DOI: 10.3389/fcell.2020.569454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) provides cues to direct mammogenesis, tumourigenesis and metastatic processes. Over the past several decades, two-dimensional (2D) culture models have been invaluable in furthering our understanding of the tumor microenvironment (TME), however, they still do not accurately emulate the associated biological complexities. In contrast, three-dimensional (3D) culture models provide a more physiologically relevant platform to study relevant physicochemical signals, stromal-epithelial cell interactions, vascular and immune components, and cell-ECM interactions in the human breast microenvironment. A common thread that may weave these multiple interactions are the proteoglycans (PGs), a prominent family of molecules in breast tissue. This review will discuss how these PGs contribute to the breast cancer TME and provide a summary of the traditional and emerging technologies that have been utilized to better understand the role of PGs during malignant transformation. Furthermore, this review will emphasize the differences that PGs exhibit between normal tissues and tumor ECM, providing a rationale for the investigation of underexplored roles of PGs in breast cancer progression using state-of-the-art 3D culture models.
Collapse
Affiliation(s)
- Julien Clegg
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Maria K Koch
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| | - Larisa M Haupt
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia.,Centre for Genomics and Personalized Health, Genomics Research Centre, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Priyakshi Kalita-de Croft
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Herston, QLD, Australia
| | - Laura J Bray
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia.,Faculty of Science and Engineering, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|