1
|
Zhao L, Gui Y, Deng X. Focus on mechano-immunology: new direction in cancer treatment. Int J Surg 2025; 111:2590-2602. [PMID: 39764598 DOI: 10.1097/js9.0000000000002224] [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: 07/31/2024] [Accepted: 11/29/2024] [Indexed: 03/16/2025]
Abstract
The immune response is modulated by a diverse array of signals within the tissue microenvironment, encompassing biochemical factors, mechanical forces, and pressures from adjacent tissues. Furthermore, the extracellular matrix and its constituents significantly influence the function of immune cells. In the case of carcinogenesis, changes in the biophysical properties of tissues can impact the mechanical signals received by immune cells, and these signals c1an be translated into biochemical signals through mechano-transduction pathways. These mechano-transduction pathways have a profound impact on cellular functions, influencing processes such as cell activation, metabolism, proliferation, and migration, etc. Tissue mechanics may undergo temporal changes during the process of carcinogenesis, offering the potential for novel dynamic levels of immune regulation. Here, we review advances in mechanoimmunology in malignancy studies, focusing on how mechanosignals modulate the behaviors of immune cells at the tissue level, thereby triggering an immune response that ultimately influences the development and progression of malignant tumors. Additionally, we have also focused on the development of mechano-immunoengineering systems, with the help of which could help to further understand the response of tumor cells or immune cells to alterations in the microenvironment and may provide new research directions for overcoming immunotherapeutic resistance of malignant tumors.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
| | - Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
| | - Xiangying Deng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Kolaczkowski OM, Goodson BA, Vazquez VM, Jia J, Bhat AQ, Kim TH, Pu J. Synergistic Role of Amino Acids in Enhancing mTOR Activation Through Lysosome Positioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.618047. [PMID: 39416115 PMCID: PMC11482915 DOI: 10.1101/2024.10.12.618047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Lysosome positioning, or lysosome cellular distribution, is critical for lysosomal functions in response to both extracellular and intracellular cues. Amino acids, as essential nutrients, have been shown to promote lysosome movement toward the cell periphery. Peripheral lysosomes are involved in processes such as lysosomal exocytosis, cell migration, and metabolic signaling-functions that are particularly important for cancer cell motility and growth. However, the specific types of amino acids that regulate lysosome positioning, their underlying mechanisms, and their connection to amino acid-regulated metabolic signaling remain poorly understood. In this study, we developed a high-content imaging system for unbiased, quantitative analysis of lysosome positioning. We examined the 15 amino acids present in cell culture media and found that 10 promoted lysosome redistribution toward the cell periphery to varying extents, with aromatic amino acids showing the strongest effect. This redistribution was mediated by promoting outward transport through SLC38A9-BORC-kinesin 1/3 axis and simultaneously reducing inward transport via inhibiting the recruitment of Rab7 and JIP4 onto lysosomes. When examining the effects of amino acids on mTOR activation-a central regulator of cell metabolism-we found that the amino acids most strongly promoting lysosome dispersal, such as phenylalanine, did not activate mTOR on their own. However, combining phenylalanine with arginine, which activates mTOR without affecting lysosome positioning, synergistically enhanced mTOR activity. This synergy was lost when lysosomes failed to localize to the cell periphery, as observed in kinesin 1/3 knockout (KO) cells. Furthermore, breast cancer cells exhibited heightened sensitivity to phenylalanine-induced lysosome dispersal compared to noncancerous breast cells. Inhibition of LAT1, the amino acid transporter responsible for phenylalanine uptake, reduced peripheral lysosomes and impaired cancer cell migration and proliferation, highlighting the importance of lysosome positioning in these coordinated cellular activities. In summary, amino acid-regulated lysosome positioning and mTOR signaling depend on distinct sets of amino acids. Combining lysosome-dispersing amino acids with mTOR-activating amino acids synergistically enhances mTOR activation, which may be particularly relevant in cancer cells.
Collapse
Affiliation(s)
- Oralia M. Kolaczkowski
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Baley A. Goodson
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Valeria Montenegro Vazquez
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Jingyue Jia
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Aadil Qadir Bhat
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Tae-Hyung Kim
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Jing Pu
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
3
|
Zhou S, Xu H, Duan Y, Tang Q, Huang H, Bi F. Survival mechanisms of circulating tumor cells and their implications for cancer treatment. Cancer Metastasis Rev 2024; 43:941-957. [PMID: 38436892 DOI: 10.1007/s10555-024-10178-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Metastasis remains the principal trigger for relapse and mortality across diverse cancer types. Circulating tumor cells (CTCs), which originate from the primary tumor or its metastatic sites, traverse the vascular system, serving as precursors in cancer recurrence and metastasis. Nevertheless, before CTCs can establish themselves in the distant parenchyma, they must overcome significant challenges present within the circulatory system, including hydrodynamic shear stress (HSS), oxidative damage, anoikis, and immune surveillance. Recently, there has been a growing body of compelling evidence suggesting that a specific subset of CTCs can persist within the bloodstream, but the precise mechanisms of their survival remain largely elusive. This review aims to present an outline of the survival challenges encountered by CTCs and to summarize the recent advancements in understanding the underlying survival mechanisms, suggesting their implications for cancer treatment.
Collapse
Affiliation(s)
- Shuang Zhou
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huanji Xu
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yichun Duan
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qiulin Tang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huixi Huang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Feng Bi
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
4
|
Scott OW, TinTin S, Cavadino A, Elwood JM. Beta-blocker use and breast cancer outcomes: a meta-analysis. Breast Cancer Res Treat 2024; 206:443-463. [PMID: 38837086 PMCID: PMC11208256 DOI: 10.1007/s10549-024-07263-4] [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: 11/08/2023] [Accepted: 01/18/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE Beta blockers (BBs) are commonly used cardiovascular medications, and their association with breast cancer outcomes has been examined in several previous observational studies and meta-analyses. In this study, an updated meta-analysis was undertaken to ascertain the association between BBs and both breast cancer death (BCD) and breast cancer recurrence (BCR). METHODS Articles were sourced from various databases up until the 14th of August 2023. Effect estimates were pooled using the random effects model, and the Higgins I2 statistic was computed to ascertain heterogeneity. Subgroup analyses were conducted by the potential for immortal time bias (ITB), the exposure period (prediagnosis vs postdiagnosis), and type of BB (selective vs non-selective). Publication bias was assessed using funnel plots and Egger's regression tests. RESULTS Twenty-four studies were included. Pooled results showed that there was no statistically significant association between BB use and both BCD (19 studies, hazard ratio = 0.90, 95% CI 0.78-1.04) and BCR (16 studies, HR = 0.87, 95% CI 0.71-1.08). After removing studies with ITB, the associations were attenuated towards the null. There was no effect modification for either outcome when stratifying by the exposure period or type of BB. There was clear evidence of publication bias for both outcomes. CONCLUSION In this meta-analysis, we found no evidence of an association between BB use and both BCD and BCR. Removing studies with ITB attenuated the associations towards the null, but there was no effect modification by the exposure period or type of BB.
Collapse
Affiliation(s)
- Oliver William Scott
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| | - Sandar TinTin
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Alana Cavadino
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - J Mark Elwood
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Building 507, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
5
|
Liang L, Song X, Zhao H, Lim CT. Insights into the mechanobiology of cancer metastasis via microfluidic technologies. APL Bioeng 2024; 8:021506. [PMID: 38841688 PMCID: PMC11151435 DOI: 10.1063/5.0195389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
During cancer metastasis, cancer cells will encounter various microenvironments with diverse physical characteristics. Changes in these physical characteristics such as tension, stiffness, viscosity, compression, and fluid shear can generate biomechanical cues that affect cancer cells, dynamically influencing numerous pathophysiological mechanisms. For example, a dense extracellular matrix drives cancer cells to reorganize their cytoskeleton structures, facilitating confined migration, while this dense and restricted space also acts as a physical barrier that potentially results in nuclear rupture. Identifying these pathophysiological processes and understanding their underlying mechanobiological mechanisms can aid in the development of more effective therapeutics targeted to cancer metastasis. In this review, we outline the advances of engineering microfluidic devices in vitro and their role in replicating tumor microenvironment to mimic in vivo settings. We highlight the potential cellular mechanisms that mediate their ability to adapt to different microenvironments. Meanwhile, we also discuss some important mechanical cues that still remain challenging to replicate in current microfluidic devices in future direction. While much remains to be explored about cancer mechanobiology, we believe the developments of microfluidic devices will reveal how these physical cues impact the behaviors of cancer cells. It will be crucial in the understanding of cancer metastasis, and potentially contributing to better drug development and cancer therapy.
Collapse
Affiliation(s)
- Lanfeng Liang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Xiao Song
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | | |
Collapse
|
6
|
Wu Y, Cao Y, Chen L, Lai X, Zhang S, Wang S. Role of Exosomes in Cancer and Aptamer-Modified Exosomes as a Promising Platform for Cancer Targeted Therapy. Biol Proced Online 2024; 26:15. [PMID: 38802766 PMCID: PMC11129508 DOI: 10.1186/s12575-024-00245-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Exosomes are increasingly recognized as important mediators of intercellular communication in cancer biology. Exosomes can be derived from cancer cells as well as cellular components in tumor microenvironment. After secretion, the exosomes carrying a wide range of bioactive cargos can be ingested by local or distant recipient cells. The released cargos act through a variety of mechanisms to elicit multiple biological effects and impact most if not all hallmarks of cancer. Moreover, owing to their excellent biocompatibility and capability of being easily engineered or modified, exosomes are currently exploited as a promising platform for cancer targeted therapy. In this review, we first summarize the current knowledge of roles of exosomes in risk and etiology, initiation and progression of cancer, as well as their underlying molecular mechanisms. The aptamer-modified exosome as a promising platform for cancer targeted therapy is then briefly introduced. We also discuss the future directions for emerging roles of exosome in tumor biology and perspective of aptamer-modified exosomes in cancer therapy.
Collapse
Affiliation(s)
- Yating Wu
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Medical Oncology, Fuzhou General Clinical Medical School (the 900th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Yue Cao
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Li Chen
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Xiaofeng Lai
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China
| | - Shenghang Zhang
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China.
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China.
| | - Shuiliang Wang
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, Fujian Province, P. R. China.
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (the 900 th Hospital), Fujian Medical University, Fujian Province, Fuzhou, P. R. China.
| |
Collapse
|
7
|
Gonzalez-Llerena JL, Espinosa-Rodriguez BA, Treviño-Almaguer D, Mendez-Lopez LF, Carranza-Rosales P, Gonzalez-Barranco P, Guzman-Delgado NE, Romo-Mancillas A, Balderas-Renteria I. Cordycepin Triphosphate as a Potential Modulator of Cellular Plasticity in Cancer via cAMP-Dependent Pathways: An In Silico Approach. Int J Mol Sci 2024; 25:5692. [PMID: 38891880 PMCID: PMC11171877 DOI: 10.3390/ijms25115692] [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: 04/05/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Cordycepin, or 3'-deoxyadenosine, is an adenosine analog with a broad spectrum of biological activity. The key structural difference between cordycepin and adenosine lies in the absence of a hydroxyl group at the 3' position of the ribose ring. Upon administration, cordycepin can undergo an enzymatic transformation in specific tissues, forming cordycepin triphosphate. In this study, we conducted a comprehensive analysis of the structural features of cordycepin and its derivatives, contrasting them with endogenous purine-based metabolites using chemoinformatics and bioinformatics tools in addition to molecular dynamics simulations. We tested the hypothesis that cordycepin triphosphate could bind to the active site of the adenylate cyclase enzyme. The outcomes of our molecular dynamics simulations revealed scores that are comparable to, and superior to, those of adenosine triphosphate (ATP), the endogenous ligand. This interaction could reduce the production of cyclic adenosine monophosphate (cAMP) by acting as a pseudo-ATP that lacks a hydroxyl group at the 3' position, essential to carry out nucleotide cyclization. We discuss the implications in the context of the plasticity of cancer and other cells within the tumor microenvironment, such as cancer-associated fibroblast, endothelial, and immune cells. This interaction could awaken antitumor immunity by preventing phenotypic changes in the immune cells driven by sustained cAMP signaling. The last could be an unreported molecular mechanism that helps to explain more details about cordycepin's mechanism of action.
Collapse
Affiliation(s)
- Jose Luis Gonzalez-Llerena
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
- Center for Research on Nutrition and Public Health, School of Public Health and Nutrition, Autonomous University of Nuevo Leon, Monterrey 66460, Mexico;
| | - Bryan Alejandro Espinosa-Rodriguez
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Daniela Treviño-Almaguer
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Luis Fernando Mendez-Lopez
- Center for Research on Nutrition and Public Health, School of Public Health and Nutrition, Autonomous University of Nuevo Leon, Monterrey 66460, Mexico;
| | - Pilar Carranza-Rosales
- Laboratory of Cell Biology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Mexico;
| | - Patricia Gonzalez-Barranco
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| | - Nancy Elena Guzman-Delgado
- Health Research Division, High Specialty Medical Unit, Cardiology Hospital N. 34. Mexican Social Security Institute, Monterrey 64360, Mexico;
| | - Antonio Romo-Mancillas
- Computer Aided Drug Design and Synthesis Group, School of Chemistry, Autonomous University of Queretaro, Queretaro 76010, Mexico
| | - Isaias Balderas-Renteria
- Laboratory of Molecular Pharmacology and Biological Models, School of Chemistry, Autonomous University of Nuevo Leon, San Nicolas de los Garza 66451, Mexico; (J.L.G.-L.); (B.A.E.-R.); (D.T.-A.); (P.G.-B.)
| |
Collapse
|
8
|
Barnard ME, Wang X, Petrick JL, Zirpoli GR, Jones D, Johnson WE, Palmer JR. Psychosocial stressors and breast cancer gene expression in the Black Women's Health Study. Breast Cancer Res Treat 2024; 204:327-340. [PMID: 38127176 PMCID: PMC11232497 DOI: 10.1007/s10549-023-07182-w] [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: 09/22/2023] [Accepted: 11/03/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE Prior studies indicate that the physiologic response to stress can affect gene expression. We evaluated differential gene expression in breast cancers collected from Black women with high versus low exposure to psychosocial stressors. METHODS We analyzed tumor RNA sequencing data from 417 Black Women's Health Study breast cancer cases with data on early life trauma and neighborhood disadvantage. We conducted age-adjusted differential gene expression analyses and pathway analyses. We also evaluated Conserved Transcriptional Response to Adversity (CTRA) contrast scores, relative fractions of immune cell types, T cell exhaustion, and adrenergic signaling. Analyses were run separately for estrogen receptor positive (ER+; n = 299) and ER- (n = 118) cases. RESULTS Among ER+ cases, the top differentially expressed pathways by stress exposure were related to RNA and protein metabolism. Among ER- cases, they were related to developmental biology, signal transduction, metabolism, and the immune system. Targeted analyses indicated greater immune pathway enrichment with stress exposure for ER- cases, and possible relevance of adrenergic signaling for ER+ cases. CTRA contrast scores did not differ by stress exposure, but in analyses of the CTRA components, ER- breast cancer cases with high neighborhood disadvantage had higher pro-inflammatory gene expression (p = 0.039) and higher antibody gene expression (p = 0.006) compared to those with low neighborhood disadvantage. CONCLUSION There are multiple pathways through which psychosocial stress exposure may influence breast tumor biology. Given the present findings on inflammation and immune response in ER- tumors, further research to identify stress-induced changes in the etiology and progression of ER- breast cancer is warranted.
Collapse
Affiliation(s)
- Mollie E Barnard
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord St., Boston, MA, 02118, USA
| | - Xutao Wang
- Division of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Jessica L Petrick
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord St., Boston, MA, 02118, USA
| | - Gary R Zirpoli
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord St., Boston, MA, 02118, USA
| | - Dennis Jones
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - W Evan Johnson
- Division of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Division of Infectious Disease, Center for Data Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Julie R Palmer
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord St., Boston, MA, 02118, USA.
| |
Collapse
|
9
|
Nan J, Roychowdhury S, Randles A. Investigating the Influence of Heterogeneity Within Cell Types on Microvessel Network Transport. Cell Mol Bioeng 2023; 16:497-507. [PMID: 38099216 PMCID: PMC10716099 DOI: 10.1007/s12195-023-00790-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Background Current research on the biophysics of circulating tumor cells often overlooks the heterogeneity of cell populations, focusing instead on average cellular properties. This study aims to address the gap by considering the diversity of cell biophysical characteristics and their implications on cancer spread. Methods We utilized computer simulations to assess the influence of variations in cell size and membrane elasticity on the behavior of cells within fluid environments. The study controlled cell and fluid properties to systematically investigate the transport of tumor cells through a simulated network of branching channels. Results The simulations revealed that even minor differences in cellular properties, such as slight changes in cell radius or shear elastic modulus, lead to significant changes in the fluid conditions that cells experience, including velocity and wall shear stress (p < 0.001). Conclusion The findings underscore the importance of considering cell heterogeneity in biophysical studies and suggest that small variations in cellular characteristics can profoundly impact the dynamics of tumor cell circulation. This has potential implications for understanding the mechanisms of cancer metastasis and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Junyu Nan
- Department of Biomedical Engineering, Duke University, Durham, USA
| | | | - Amanda Randles
- Department of Biomedical Engineering, Duke University, Durham, USA
| |
Collapse
|
10
|
Kwon S, Han SJ, Kim KS. Differential response of MDA‑MB‑231 breast cancer and MCF10A normal breast cells to cytoskeletal disruption. Oncol Rep 2023; 50:200. [PMID: 37772386 PMCID: PMC10565893 DOI: 10.3892/or.2023.8637] [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: 03/31/2023] [Accepted: 07/27/2023] [Indexed: 09/30/2023] Open
Abstract
Metastasis remains a major clinical problem in cancer diagnosis and treatment. Metastasis is the leading cause of cancer‑related mortality but is still poorly understood. Cytoskeletal proteins are considered potential therapeutic targets for metastatic cancer cells because the cytoskeleton serves a key role in the migration and invasion of these cells. Vimentin and F‑actin exhibit several functional similarities and undergo quantitative and structural changes during carcinogenesis. The present study investigated the effects of vimentin and F‑actin deficiency on the survival and motility of breast cancer cells. In metastatic breast cancer cells (MDA‑MB‑231) and breast epithelial cells (MCF10A), vimentin was knocked down by small interfering RNA and F‑actin was depolymerized by latrunculin A, respectively. The effect of reduced vimentin and F‑actin content on cell viability was analyzed using the MTT assay and the proliferative capacity was compared by analyzing the recovery rate. The effect on motility was analyzed based on two processes: The distance traveled by tracking the cell nucleus and the movement of the protrusions. The effects on cell elasticity were measured using atomic force microscopy. Separately reducing vimentin or F‑actin did not effectively inhibit the growth and motility of MDA‑MB‑231 cells; however, when both vimentin and F‑actin were simultaneously deficient, MDA‑MB‑231 cells growth and migration were severely impaired. Vimentin deficiency in MDA‑MB‑231 cells was compensated by an increase in F‑actin polymerization, but no complementary action of vimentin on the decrease in F‑actin was observed. In MCF10A cells, no complementary interaction was observed for both vimentin and F‑actin.
Collapse
Affiliation(s)
- Sangwoo Kwon
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Se Jik Han
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
11
|
Lempesis IG, Georgakopoulou VE, Papalexis P, Chrousos GP, Spandidos DA. Role of stress in the pathogenesis of cancer (Review). Int J Oncol 2023; 63:124. [PMID: 37711028 PMCID: PMC10552722 DOI: 10.3892/ijo.2023.5572] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023] Open
Abstract
Stress is a state of disrupted homeostasis, triggered by intrinsic or extrinsic factors, the stressors, which are counteracted by various physiological and behavioural adaptive responses. Stress has been linked to cancer development and incidence for decades; however, epidemiological studies and clinical trials have yielded contradictory results. The present review discusses the effects of stress on cancer development and the various underlying mechanisms. Animal studies have revealed a clear link between stress and cancer progression, revealing molecular, cellular and endocrine processes that are implicated in these effects. Thus, stress hormones, their receptor systems and their intracellular molecular pathways mediate the effects of stress on cancer initiation, progression and the development of metastases. The mechanisms linking stress and cancer progression can either be indirect, mediated by changes in the cancer microenvironment or immune system dysregulation, or direct, through the binding of neuroendocrine stress‑related signalling molecules to cancer cell receptors. Stress affects numerous anti‑ and pro‑cancer immune system components, including host resistance to metastasis, tumour retention and/or immune suppression. Chronic psychological stress through the elevation of catecholamine levels may increase cancer cell death resistance. On the whole, stress is linked to cancer development and incidence, with psychological stressors playing a crucial role. Animal studies have revealed a better link than human ones, with stress‑related hormones influencing tumour development, migration, invasion and cell proliferation. Randomized controlled trials are required to further evaluate the long‑term cancer outcomes of stress and its management.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Pathophysiology, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vasiliki Epameinondas Georgakopoulou
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Pathophysiology, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Petros Papalexis
- Unit of Endocrinology, First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | - Georgios P. Chrousos
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
- University Research Institute of Maternal and Child Health and Precision Medicine and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, 11527 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
12
|
Herrera-Reinoza N, Tortelli Junior TC, Teixeira FDS, Chammas R, Salvadori MC. Role of galectin-3 in the elastic response of radial growth phase melanoma cancer cells. Microsc Res Tech 2023; 86:1353-1362. [PMID: 37070727 DOI: 10.1002/jemt.24328] [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: 09/14/2022] [Revised: 02/28/2023] [Accepted: 04/06/2023] [Indexed: 04/19/2023]
Abstract
Melanoma is originated from the malignant transformation of the melanocytes and is characterized by a high rate of invasion, the more serious stage compromising deeper layers of the skin and eventually leading to the metastasis. A high mortality due to melanoma lesion persists because most of melanoma lesions are detected in advanced stages, which decreases the chances of survival. The identification of the principal mechanics implicated in the development and progression of melanoma is essential to devise new early diagnosis strategies. Cell mechanics is related with a lot of cellular functions and processes, for instance motility, differentiation, migration and invasion. In particular, the elastic modulus (Young's modulus) is a very explored parameter to describe the cell mechanical properties; most cancer cells reported in the literature smaller elasticity modulus. In this work, we show that the elastic modulus of melanoma cells lacking galectin-3 is significantly lower than those of melanoma cells expressing galectin-3. More interestingly, the gradient of elastic modulus in cells from the nuclear region towards the cell periphery is more pronounced in shGal3 cells. RESEARCH HIGHLIGHTS: AFM imaging and force spectroscopy were used to investigate the morphology and elasticity properties of healthy HaCaT cells and melanoma cells WM1366, with (shSCR) and without (shGal3) expression of galectin-3. It is shown the effect of galectin-3 protein on the elastic properties of cells: the cells without expression of galectin-3 presents lower elastic modulus. By the results, we suggest here that galectin-3 could be used as an effective biomarker of malignancy in both melanoma diagnostic and prognosis.
Collapse
Affiliation(s)
| | | | | | - Roger Chammas
- Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
13
|
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
|
14
|
Oh M, Batty S, Banerjee N, Kim TH. High extracellular glucose promotes cell motility by modulating cell deformability and contractility via the cAMP-RhoA-ROCK axis in human breast cancer cells. Mol Biol Cell 2023; 34:ar79. [PMID: 37195739 PMCID: PMC10398875 DOI: 10.1091/mbc.e22-12-0560] [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: 12/19/2022] [Revised: 04/18/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023] Open
Abstract
The mechanical properties, or mechanotypes, of cells are largely determined by their deformability and contractility. The ability of cancer cells to deform and generate contractile force is critical in multiple steps of metastasis. Identifying soluble cues that regulate cancer cell mechanotypes and understanding the underlying molecular mechanisms regulating these cellular mechanotypes could provide novel therapeutic targets to prevent metastasis. Although a strong correlation between high glucose level and cancer metastasis has been demonstrated, the causality has not been elucidated, and the underlying molecular mechanisms remain largely unknown. In this study, using novel high-throughput mechanotyping assays, we show that human breast cancer cells become less deformable and more contractile with increased extracellular glucose levels (>5 mM). These altered cell mechanotypes are due to increased F-actin rearrangement and nonmuscle myosin II (NMII) activity. We identify the cAMP-RhoA-ROCK-NMII axis as playing a major role in regulating cell mechanotypes at high extracellular glucose levels, whereas calcium and myosin light-chain kinase (MLCK) are not required. The altered mechanotypes are also associated with increased cell migration and invasion. Our study identifies key components in breast cancer cells that convert high extracellular glucose levels into changes in cellular mechanotype and behavior relevant in cancer metastasis.
Collapse
Affiliation(s)
- Mijung Oh
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Skylar Batty
- Undergraduate Pipeline Network Summer Research Program, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721
| | - Nayan Banerjee
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, West Bengal, India
| | - Tae-Hyung Kim
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131
| |
Collapse
|
15
|
Du J, Rui F, Hao Z, Hang Y, Shu J. Transcription Factor E2F1 Regulates the Expression of ADRB2. Int J Anal Chem 2023; 2023:8210685. [PMID: 37128280 PMCID: PMC10148742 DOI: 10.1155/2023/8210685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 02/09/2023] [Accepted: 02/24/2023] [Indexed: 05/03/2023] Open
Abstract
Adrenergic beta-2-receptor (ADRB2) is highly expressed in various tissue cells, affecting the susceptibility, development, and drug efficacy of diseases such as bronchial asthma and malignant tumor. However, the transcriptional regulatory mechanism of the human ADRB2 gene remains unclear. This study aimed to clarify whether E2F transcription factor 1 (E2F1) was involved in the transcriptional regulation of the human ADRB2 gene. First, the 5' flanking region of the human ADRB2 gene was cloned, and its activity was detected using A549 and BEAS-2B cells. Second, it was found that the overexpression of E2F1 could increase promoter activity by a dual-luciferase reporter gene assay. In contrast, treatment of knockdown of E2F1 significantly resulted in a decrease in its promoter activity. Moreover, mutation of the binding site of E2F1 greatly reduced the potential of human ADRB2 promoter transcriptional activity to be regulated by E2F1 overexpression and knockdown. Additionally, by real-time quantitative PCR and Western blot analysis, we demonstrated that overexpression of E2F1 elevated the ADRB2 mRNA expression and protein levels while si-E2F1 reduced its expression. Finally, the consequence of the chromatin immunoprecipitation assay showed that E2F1 was able to bind to the promoter region of ADRB2 in vivo. These results confirmed that E2F1 upregulated the expression of the human ADRB2 gene.
Collapse
Affiliation(s)
- Juan Du
- Department of Pediatrics, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feifei Rui
- Department of Neonatology, Changzhou Maternal and Child Health Hospital, Changzhou, Jiangsu, China
| | - Zhongfen Hao
- Department of Pediatrics, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun Hang
- Department of Pediatrics, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jin Shu
- Department of Pediatrics, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
16
|
Abstract
Immune responses are governed by signals from the tissue microenvironment, and in addition to biochemical signals, mechanical cues and forces arising from the tissue, its extracellular matrix and its constituent cells shape immune cell function. Indeed, changes in biophysical properties of tissue alter the mechanical signals experienced by cells in many disease conditions, in inflammatory states and in the context of ageing. These mechanical cues are converted into biochemical signals through the process of mechanotransduction, and multiple pathways of mechanotransduction have been identified in immune cells. Such pathways impact important cellular functions including cell activation, cytokine production, metabolism, proliferation and trafficking. Changes in tissue mechanics may also represent a new form of 'danger signal' that alerts the innate and adaptive immune systems to the possibility of injury or infection. Tissue mechanics can change temporally during an infection or inflammatory response, offering a novel layer of dynamic immune regulation. Here, we review the emerging field of mechanoimmunology, focusing on how mechanical cues at the scale of the tissue environment regulate immune cell behaviours to initiate, propagate and resolve the immune response.
Collapse
|
17
|
Sivagurunathan S, Vahabikashi A, Yang H, Zhang J, Vazquez K, Rajasundaram D, Politanska Y, Abdala-Valencia H, Notbohm J, Guo M, Adam SA, Goldman RD. Expression of vimentin alters cell mechanics, cell-cell adhesion, and gene expression profiles suggesting the induction of a hybrid EMT in human mammary epithelial cells. Front Cell Dev Biol 2022; 10:929495. [PMID: 36200046 PMCID: PMC9527304 DOI: 10.3389/fcell.2022.929495] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Vimentin is a Type III intermediate filament (VIF) cytoskeletal protein that regulates the mechanical and migratory behavior of cells. Its expression is considered to be a marker for the epithelial to mesenchymal transition (EMT) that takes place in tumor metastasis. However, the molecular mechanisms regulated by the expression of vimentin in the EMT remain largely unexplored. We created MCF7 epithelial cell lines expressing vimentin from a cumate-inducible promoter to address this question. When vimentin expression was induced in these cells, extensive cytoplasmic VIF networks were assembled accompanied by changes in the organization of the endogenous keratin intermediate filament networks and disruption of desmosomes. Significant reductions in intercellular forces by the cells expressing VIFs were measured by quantitative monolayer traction force and stress microscopy. In contrast, laser trapping micro-rheology revealed that the cytoplasm of MCF7 cells expressing VIFs was stiffer than the uninduced cells. Vimentin expression activated transcription of genes involved in pathways responsible for cell migration and locomotion. Importantly, the EMT related transcription factor TWIST1 was upregulated only in wild type vimentin expressing cells and not in cells expressing a mutant non-polymerized form of vimentin, which only formed unit length filaments (ULF). Taken together, our results suggest that vimentin expression induces a hybrid EMT correlated with the upregulation of genes involved in cell migration.
Collapse
Affiliation(s)
- Suganya Sivagurunathan
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Amir Vahabikashi
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Haiqian Yang
- Department of Mechanical Engineering , Massachusetts Institute of Technology , Cambridge , MA, United States
| | - Jun Zhang
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Kelly Vazquez
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yuliya Politanska
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hiam Abdala-Valencia
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jacob Notbohm
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Ming Guo
- Department of Mechanical Engineering , Massachusetts Institute of Technology , Cambridge , MA, United States
| | - Stephen A Adam
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Robert D Goldman
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
18
|
Løfling LL, Støer NC, Sloan EK, Chang A, Gandini S, Ursin G, Botteri E. β-blockers and breast cancer survival by molecular subtypes: a population-based cohort study and meta-analysis. Br J Cancer 2022; 127:1086-1096. [PMID: 35725814 PMCID: PMC9470740 DOI: 10.1038/s41416-022-01891-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 01/17/2023] Open
Abstract
Background The association between use of β-blockers and breast cancer (BC) prognosis has been investigated in several observational studies, with conflicting results. We performed a nationwide cohort study and a meta-analysis to investigate the association, and assess if it varied between molecular subtypes of BC. Methods We identified women aged ≥50 years with BC diagnosed between 2004 and 2018 in Norway. We used Cox regression models to estimate the association between β-blocker use at diagnosis and BC-specific survival, overall and by molecular subtype. We performed a meta-analysis of observational studies that reported molecular subtype-specific estimates of this association. Results We included 30,060 women, of which 4461 (15%) used β-blockers. After a median follow-up of 5.1 years, 2826 (9%) died of BC. Overall, β-blocker use was not associated with BC-specific survival (hazard ratio [HR] = 1.07; 95% confidence interval [CI]: 0.97–1.19). We found an association only in triple-negative BC (TNBC) patients (HR = 0.66; 95% CI: 0.47–0.91). This was confirmed in the meta-analysis: β-blocker use was associated with progression/recurrence-free (HR = 0.58; 95% CI: 0.38–0.89) and BC-specific survival (HR = 0.74; 95% CI: 0.55–1.00) in TNBC patients only. Conclusion In our cohort of BC patients and in the meta-analysis, β-blocker use was associated with prolonged BC-specific survival only in TNBC patients.
Collapse
Affiliation(s)
- L Lukas Løfling
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Nathalie C Støer
- Department of Research, Cancer Registry of Norway, Oslo, Norway.,Norwegian Research Centre for Women's Health, Women's Clinic, Oslo University Hospital, Oslo, Norway
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Science, Monash University, Parkville, VIC, Australia
| | - Aeson Chang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Science, Monash University, Parkville, VIC, Australia
| | - Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giske Ursin
- Cancer Registry of Norway, Oslo, Norway.,Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Edoardo Botteri
- Department of Research, Cancer Registry of Norway, Oslo, Norway. .,Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway.
| |
Collapse
|
19
|
Silva D, Quintas C, Gonçalves J, Fresco P. Contribution of adrenergic mechanisms for the stress-induced breast cancer carcinogenesis. J Cell Physiol 2022; 237:2107-2127. [PMID: 35243626 DOI: 10.1002/jcp.30707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 02/12/2022] [Indexed: 12/11/2022]
Abstract
Breast cancer is the most common and deadliest type of cancer in women. Stress exposure has been associated with carcinogenesis and the stress released neurotransmitters, noradrenaline and adrenaline, and their cognate receptors, can participate in the carcinogenesis process, either by regulating tumor microenvironment or by promoting systemic changes. This work intends to provide an overview of the research done in this area and try to unravel the role of adrenergic ligands in the context of breast carcinogenesis. In the initiation phase, adrenergic signaling may favor neoplastic transformation of breast epithelial cells whereas, during cancer progression, may favor the metastatic potential of breast cancer cells. Additionally, adrenergic signaling can alter the function and activity of other cells present in the tumor microenvironment towards a protumor phenotype, namely macrophages, fibroblasts, and by altering adipocyte's function. Adrenergic signaling also promotes angiogenesis and lymphangiogenesis and, systemically, may induce the formation of preneoplastic niches, cancer-associated cachexia and alterations in the immune system which contribute for the loss of quality of life of breast cancer patients and their capacity to fight cancer. Most studies points to a major contribution of β2 -adrenoceptor activated pathways on these effects. The current knowledge of the mechanistic pathways activated by β2 -adrenoceptors in physiology and pathophysiology, the availability of selective drugs approved for clinical use and a deeper knowledge of the basic cellular and molecular pathways by which adrenergic stimulation may influence cancer initiation and progression, opens the possibility to use new therapeutic alternatives to improve efficacy of breast cancer treatments.
Collapse
Affiliation(s)
- Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Clara Quintas
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
20
|
Angstadt S, Zhu Q, Jaffee EM, Robinson DN, Anders RA. Pancreatic Ductal Adenocarcinoma Cortical Mechanics and Clinical Implications. Front Oncol 2022; 12:809179. [PMID: 35174086 PMCID: PMC8843014 DOI: 10.3389/fonc.2022.809179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/05/2022] [Indexed: 12/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest cancers due to low therapeutic response rates and poor prognoses. Majority of patients present with symptoms post metastatic spread, which contributes to its overall lethality as the 4th leading cause of cancer-related deaths. Therapeutic approaches thus far target only one or two of the cancer specific hallmarks, such as high proliferation rate, apoptotic evasion, or immune evasion. Recent genomic discoveries reveal that genetic heterogeneity, early micrometastases, and an immunosuppressive tumor microenvironment contribute to the inefficacy of current standard treatments and specific molecular-targeted therapies. To effectively combat cancers like PDAC, we need an innovative approach that can simultaneously impact the multiple hallmarks driving cancer progression. Here, we present the mechanical properties generated by the cell’s cortical cytoskeleton, with a spotlight on PDAC, as an ideal therapeutic target that can concurrently attack multiple systems driving cancer. We start with an introduction to cancer cell mechanics and PDAC followed by a compilation of studies connecting the cortical cytoskeleton and mechanical properties to proliferation, metastasis, immune cell interactions, cancer cell stemness, and/or metabolism. We further elaborate on the implications of these findings in disease progression, therapeutic resistance, and clinical relapse. Manipulation of the cancer cell’s mechanical system has already been shown to prevent metastasis in preclinical models, but it has greater potential for target exploration since it is a foundational property of the cell that regulates various oncogenic behaviors.
Collapse
Affiliation(s)
- Shantel Angstadt
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Qingfeng Zhu
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elizabeth M. Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Douglas N. Robinson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Douglas N. Robinson, ; Robert A. Anders,
| | - Robert A. Anders
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Douglas N. Robinson, ; Robert A. Anders,
| |
Collapse
|
21
|
Zhang H, Chen Z, Zhang A, Gupte AA, Hamilton DJ. The Role of Calcium Signaling in Melanoma. Int J Mol Sci 2022; 23:ijms23031010. [PMID: 35162934 PMCID: PMC8835635 DOI: 10.3390/ijms23031010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
Calcium signaling plays important roles in physiological and pathological conditions, including cutaneous melanoma, the most lethal type of skin cancer. Intracellular calcium concentration ([Ca2+]i), cell membrane calcium channels, calcium related proteins (S100 family, E-cadherin, and calpain), and Wnt/Ca2+ pathways are related to melanogenesis and melanoma tumorigenesis and progression. Calcium signaling influences the melanoma microenvironment, including immune cells, extracellular matrix (ECM), the vascular network, and chemical and physical surroundings. Other ionic channels, such as sodium and potassium channels, are engaged in calcium-mediated pathways in melanoma. Calcium signaling serves as a promising pharmacological target in melanoma treatment, and its dysregulation might serve as a marker for melanoma prediction. We documented calcium-dependent endoplasmic reticulum (ER) stress and mitochondria dysfunction, by targeting calcium channels and influencing [Ca2+]i and calcium homeostasis, and attenuated drug resistance in melanoma management.
Collapse
Affiliation(s)
- Haoran Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.Z.); (A.Z.); (A.A.G.)
- Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Zhe Chen
- Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.Z.); (A.Z.); (A.A.G.)
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, TX 77030, USA
| | - Anisha A. Gupte
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.Z.); (A.Z.); (A.A.G.)
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, TX 77030, USA
| | - Dale J. Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA; (H.Z.); (A.Z.); (A.A.G.)
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-(713)-441-4483
| |
Collapse
|
22
|
Chang A, Sloan EK, Antoni MH, Knight JM, Telles R, Lutgendorf SK. Biobehavioral Pathways and Cancer Progression: Insights for Improving Well-Being and Cancer Outcomes. Integr Cancer Ther 2022; 21:15347354221096081. [PMID: 35579197 PMCID: PMC9118395 DOI: 10.1177/15347354221096081] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022] Open
Abstract
The relationship between psychosocial factors and cancer has intrigued people for centuries. In the last several decades there has been an expansion of mechanistic research that has revealed insights regarding how stress activates neuroendocrine stress-response systems to impact cancer progression. Here, we review emerging mechanistic findings on key pathways implicated in the effect of stress on cancer progression, including the cellular immune response, inflammation, angiogenesis, and metastasis, with a primary focus on the mediating role of the sympathetic nervous system. We discuss converging findings from preclinical and clinical cancer research that describe these pathways and research that reveals how these stress pathways may be targeted via pharmacological and mind-body based interventions. While further research is required, the body of work reviewed here highlights the need for and feasibility of an integrated approach to target stress pathways in cancer patients to achieve comprehensive cancer treatment.
Collapse
Affiliation(s)
- Aeson Chang
- Monash Institute of Pharmaceutical Sciences, Drug Discovery Biology, Monash University, Parkville, VIC, Australia
| | - Erica K. Sloan
- Monash Institute of Pharmaceutical Sciences, Drug Discovery Biology, Monash University, Parkville, VIC, Australia
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Peter MacCallum Cancer Centre, Division of Surgery, Melbourne, VIC, Australia
| | - Michael H. Antoni
- Departments of Psychology, Psychiatry, and Behavioral Sciences, and Cancer Control Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Jennifer M. Knight
- Department of Psychiatry and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Rachel Telles
- Departments of Psychological and Brain Sciences, Obstetrics and Gynecology, and Urology, and Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Susan K. Lutgendorf
- Departments of Psychological and Brain Sciences, Obstetrics and Gynecology, and Urology, and Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
23
|
Archer M, Dogra N, Dovey Z, Ganta T, Jang HS, Khusid JA, Lantz A, Mihalopoulos M, Stockert JA, Zahalka A, Björnebo L, Gaglani S, Noh MR, Kaplan SA, Mehrazin R, Badani KK, Wiklund P, Tsao K, Lundon DJ, Mohamed N, Lucien F, Padanilam B, Gupta M, Tewari AK, Kyprianou N. Role of α- and β-adrenergic signaling in phenotypic targeting: significance in benign and malignant urologic disease. Cell Commun Signal 2021; 19:78. [PMID: 34284799 PMCID: PMC8290582 DOI: 10.1186/s12964-021-00755-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 01/17/2023] Open
Abstract
The urinary tract is highly innervated by autonomic nerves which are essential in urinary tract development, the production of growth factors, and the control of homeostasis. These neural signals may become dysregulated in several genitourinary (GU) disease states, both benign and malignant. Accordingly, the autonomic nervous system is a therapeutic target for several genitourinary pathologies including cancer, voiding dysfunction, and obstructing nephrolithiasis. Adrenergic receptors (adrenoceptors) are G-Protein coupled-receptors that are distributed throughout the body. The major function of α1-adrenoceptors is signaling smooth muscle contractions through GPCR and intracellular calcium influx. Pharmacologic intervention of α-and β-adrenoceptors is routinely and successfully implemented in the treatment of benign urologic illnesses, through the use of α-adrenoceptor antagonists. Furthermore, cell-based evidence recently established the antitumor effect of α1-adrenoceptor antagonists in prostate, bladder and renal tumors by reducing neovascularity and impairing growth within the tumor microenvironment via regulation of the phenotypic epithelial-mesenchymal transition (EMT). There has been a significant focus on repurposing the routinely used, Food and Drug Administration-approved α1-adrenoceptor antagonists to inhibit GU tumor growth and angiogenesis in patients with advanced prostate, bladder, and renal cancer. In this review we discuss the current evidence on (a) the signaling events of the autonomic nervous system mediated by its cognate α- and β-adrenoceptors in regulating the phenotypic landscape (EMT) of genitourinary organs; and (b) the therapeutic significance of targeting this signaling pathway in benign and malignant urologic disease. Video abstract.
Collapse
Affiliation(s)
- M. Archer
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - N. Dogra
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Z. Dovey
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - T. Ganta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - H.-S. Jang
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - J. A. Khusid
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Lantz
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institute, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - M. Mihalopoulos
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - J. A. Stockert
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Zahalka
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - L. Björnebo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - S. Gaglani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. R. Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - S. A. Kaplan
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - R. Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. K. Badani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - P. Wiklund
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. Tsao
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - D. J. Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Mohamed
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - F. Lucien
- Department of Urology, Mayo Clinic, Rochester, MN USA
| | - B. Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. Gupta
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. K. Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
24
|
Hymel SJ, Fujioka H, Khismatullin DB. Modeling of Deformable Cell Separation in a Microchannel with Sequenced Pillars. ADVANCED THEORY AND SIMULATIONS 2021. [DOI: 10.1002/adts.202100086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Scott J. Hymel
- Department of Biomedical Engineering Tulane University New Orleans LA 70118 USA
| | - Hideki Fujioka
- Center for Computational Science Tulane University New Orleans LA 70118 USA
| | - Damir B. Khismatullin
- Department of Biomedical Engineering Tulane University New Orleans LA 70118 USA
- Center for Computational Science Tulane University New Orleans LA 70118 USA
| |
Collapse
|
25
|
Mravec B. Neurobiology of Cancer: Introduction of New Drugs in the Treatment and Prevention of Cancer. Int J Mol Sci 2021; 22:6115. [PMID: 34204103 PMCID: PMC8201304 DOI: 10.3390/ijms22116115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022] Open
Abstract
Research on the neurobiology of cancer, which lies at the border of neuroscience and oncology, has elucidated the mechanisms and pathways that enable the nervous system to modulate processes associated with cancer initiation and progression. This research has also shown that several drugs which modulate interactions between the nervous system and the tumor micro- and macroenvironments significantly reduced the progression of cancer in animal models. Encouraging results were also provided by prospective clinical trials investigating the effect of drugs that reduce adrenergic signaling on the course of cancer in oncological patients. Moreover, it has been shown that reducing adrenergic signaling might also reduce the incidence of cancer in animal models, as well as in humans. However, even if many experimental and clinical findings have confirmed the preventive and therapeutic potential of drugs that reduce the stimulatory effect of the nervous system on processes related to cancer initiation and progression, several questions remain unanswered. Therefore, the aim of this review is to critically evaluate the efficiency of these drugs and to discuss questions that need to be answered before their introduction into conventional cancer treatment and prevention.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 813 72 Bratislava, Slovakia; ; Tel.: +421-(2)-59357527; Fax: +421-(2)-59357601
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| |
Collapse
|
26
|
Lee H, Bonin K, Guthold M. Human mammary epithelial cells in a mature, stratified epithelial layer flatten and stiffen compared to single and confluent cells. Biochim Biophys Acta Gen Subj 2021; 1865:129891. [PMID: 33689830 DOI: 10.1016/j.bbagen.2021.129891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND The epithelium forms a protective barrier against external biological, chemical and physical insults. So far, AFM-based, micro-mechanical measurements have only been performed on single cells and confluent cells, but not yet on cells in mature layers. METHODS Using a combination of atomic force, fluorescence and confocal microscopy, we determined the changes in stiffness, morphology and actin distribution of human mammary epithelial cells (HMECs) as they transition from single cells to confluency to a mature layer. RESULTS Single HMECs have a tall, round (planoconvex) morphology, have actin stress fibers at the base, have diffuse cortical actin, and have a stiffness of 1 kPa. Confluent HMECs start to become flatter, basal actin stress fibers start to disappear, and actin accumulates laterally where cells abut. Overall stiffness is still 1 kPa with two-fold higher stiffness in the abutting regions. As HMECs mature and form multilayered structures, cells on apical surfaces become flatter (apically more level), wider, and seven times stiffer (mean, 7 kPa) than single and confluent cells. The main drivers of these changes are actin filaments, as cells show strong actin accumulation in the regions where cells adjoin, and in the apical regions. CONCLUSIONS HMECs stiffen, flatten and redistribute actin upon transiting from single cells to mature, confluent layers. GENERAL SIGNIFICANCE Our findings advance the understanding of breast ductal morphogenesis and mechanical homeostasis.
Collapse
Affiliation(s)
- Hyunsu Lee
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Keith Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Martin Guthold
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA.
| |
Collapse
|
27
|
The role of ADRB2 gene polymorphisms in malignancies. Mol Biol Rep 2021; 48:2741-2749. [PMID: 33675465 DOI: 10.1007/s11033-021-06250-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/24/2021] [Indexed: 12/30/2022]
Abstract
Beta-2-adrenergic receptor is a member of the G protein-coupled receptor superfamily, which is highly expressed in most malignancies. There is increasing evidence showing that beta-2-adrenergic receptors are associated with carcinogenesis, proliferation, immune regulation, invasion, angiogenesis, clinical prognosis and treatment resistance in malignancies. Polymorphisms of the ADRB2 gene have been confirmed to be associated with transcriptional activity, mRNA translation, and beta-2-adrenergic receptor expression and sensitivity. This review discusses clinically relevant examples of single nucleotide polymorphisms of ADRB2 in malignancies and the effects of these polymorphisms on cancer susceptibility, prognosis and treatment response of cancer patients.
Collapse
|
28
|
Graybill PM, Bollineni RK, Sheng Z, Davalos RV, Mirzaeifar R. A constriction channel analysis of astrocytoma stiffness and disease progression. BIOMICROFLUIDICS 2021; 15:024103. [PMID: 33763160 PMCID: PMC7968935 DOI: 10.1063/5.0040283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/23/2021] [Indexed: 05/12/2023]
Abstract
Studies have demonstrated that cancer cells tend to have reduced stiffness (Young's modulus) compared to their healthy counterparts. The mechanical properties of primary brain cancer cells, however, have remained largely unstudied. To investigate whether the stiffness of primary brain cancer cells decreases as malignancy increases, we used a microfluidic constriction channel device to deform healthy astrocytes and astrocytoma cells of grade II, III, and IV and measured the entry time, transit time, and elongation. Calculating cell stiffness directly from the experimental measurements is not possible. To overcome this challenge, finite element simulations of the cell entry into the constriction channel were used to train a neural network to calculate the stiffness of the analyzed cells based on their experimentally measured diameter, entry time, and elongation in the channel. Our study provides the first calculation of stiffness for grades II and III astrocytoma and is the first to apply a neural network analysis to determine cell mechanical properties from a constriction channel device. Our results suggest that the stiffness of astrocytoma cells is not well-correlated with the cell grade. Furthermore, while other non-central-nervous-system cell types typically show reduced stiffness of malignant cells, we found that most astrocytoma cell lines had increased stiffness compared to healthy astrocytes, with lower-grade astrocytoma having higher stiffness values than grade IV glioblastoma. Differences in nucleus-to-cytoplasm ratio only partly explain differences in stiffness values. Although our study does have limitations, our results do not show a strong correlation of stiffness with cell grade, suggesting that other factors may play important roles in determining the invasive capability of astrocytoma. Future studies are warranted to further elucidate the mechanical properties of astrocytoma across various pathological grades.
Collapse
Affiliation(s)
| | - R. K. Bollineni
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Z. Sheng
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine and Virginia Tech Fralin Biomedical Research Institute, Roanoke, Virginia 24016, USA
| | - R. V. Davalos
- Authors to whom correspondence should be addressed: and
| | - R. Mirzaeifar
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
29
|
Conceição F, Sousa DM, Paredes J, Lamghari M. Sympathetic activity in breast cancer and metastasis: partners in crime. Bone Res 2021; 9:9. [PMID: 33547275 PMCID: PMC7864971 DOI: 10.1038/s41413-021-00137-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 01/30/2023] Open
Abstract
The vast majority of patients with advanced breast cancer present skeletal complications that severely compromise their quality of life. Breast cancer cells are characterized by a strong tropism to the bone niche. After engraftment and colonization of bone, breast cancer cells interact with native bone cells to hinder the normal bone remodeling process and establish an osteolytic "metastatic vicious cycle". The sympathetic nervous system has emerged in recent years as an important modulator of breast cancer progression and metastasis, potentiating and accelerating the onset of the vicious cycle and leading to extensive bone degradation. Furthermore, sympathetic neurotransmitters and their cognate receptors have been shown to promote several hallmarks of breast cancer, such as proliferation, angiogenesis, immune escape, and invasion of the extracellular matrix. In this review, we assembled the current knowledge concerning the complex interactions that take place in the tumor microenvironment, with a special emphasis on sympathetic modulation of breast cancer cells and stromal cells. Notably, the differential action of epinephrine and norepinephrine, through either α- or β-adrenergic receptors, on breast cancer progression prompts careful consideration when designing new therapeutic options. In addition, the contribution of sympathetic innervation to the formation of bone metastatic foci is highlighted. In particular, we address the remarkable ability of adrenergic signaling to condition the native bone remodeling process and modulate the bone vasculature, driving breast cancer cell engraftment in the bone niche. Finally, clinical perspectives and developments on the use of β-adrenergic receptor inhibitors for breast cancer management and treatment are discussed.
Collapse
Affiliation(s)
- Francisco Conceição
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Daniela M. Sousa
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Paredes
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226FMUP—Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Meriem Lamghari
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
30
|
Iftikhar A, Islam M, Shepherd S, Jones S, Ellis I. Cancer and Stress: Does It Make a Difference to the Patient When These Two Challenges Collide? Cancers (Basel) 2021; 13:cancers13020163. [PMID: 33418900 PMCID: PMC7825104 DOI: 10.3390/cancers13020163] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Head and neck cancers are the sixth most common cancer in the world. The burden of the disease has remained challenging over recent years despite the advances in treatments of other malignancies. The very use of the word malignancy brings about a stress response in almost all adult patients. Being told you have a tumour is not a word anyone wants to hear. We have embarked on a study which will investigate the effect of stress pathways on head and neck cancer patients and which signalling pathways may be involved. In the future, this will allow clinicians to better manage patients with head and neck cancer and reduce the patients’ stress so that this does not add to their tumour burden. Abstract A single head and neck Cancer (HNC) is a globally growing challenge associated with significant morbidity and mortality. The diagnosis itself can affect the patients profoundly let alone the complex and disfiguring treatment. The highly important functions of structures of the head and neck such as mastication, speech, aesthetics, identity and social interactions make a cancer diagnosis in this region even more psychologically traumatic. The emotional distress engendered as a result of functional and social disruption is certain to negatively affect health-related quality of life (HRQoL). The key biological responses to stressful events are moderated through the combined action of two systems, the hypothalamus–pituitary–adrenal axis (HPA) which releases glucocorticoids and the sympathetic nervous system (SNS) which releases catecholamines. In acute stress, these hormones help the body to regain homeostasis; however, in chronic stress their increased levels and activation of their receptors may aid in the progression of cancer. Despite ample evidence on the existence of stress in patients diagnosed with HNC, studies looking at the effect of stress on the progression of disease are scarce, compared to other cancers. This review summarises the challenges associated with HNC that make it stressful and describes how stress signalling aids in the progression of cancer. Growing evidence on the relationship between stress and HNC makes it paramount to focus future research towards a better understanding of stress and its effect on head and neck cancer.
Collapse
|
31
|
Chandore H, Raj AK, Lokhande KB, Swamy KV, Pal JK, Sharma NK. An Intracellular Tripeptide Arg-His-Trp of Serum Origin Detected in MCF-7 Cells is a Possible Agonist to β2 Adrenoceptor. Protein Pept Lett 2021; 28:1191-1202. [PMID: 34397320 DOI: 10.2174/0929866528666210816114901] [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: 02/24/2021] [Revised: 05/29/2021] [Accepted: 06/08/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The need for agonists and antagonists of β2 adrenoceptor (β2AR) is warranted in various human disease conditions, including cancer, cardiovascular and other metabolic disorders. However, the sources of agonists of β2AR are diverse in nature. Interestingly, there is a complete gap in the exploration of agonists of β2AR from serum that is a well-known component of culture media that supports growth and proliferation of normal and cancer cells in vitro. METHODS In this paper, we employed a novel vertical tube gel electrophoresis (VTGE)-assisted purification of intracellular metabolites of MCF-7 cells grown in vitro in complete media with fetal bovine serum (FBS). Intracellular metabolites of MCF-7 cells were then analyzed by LC-HRMS. Identified intracellular tripeptides of FBS origin were evaluated for their molecular interactions with various extracellular and intracellular receptors, including β2AR (PDB ID: 2RH1) by employing molecular docking and molecular dynamics simulations (MDS). A known agonist of β2AR, isoproterenol was used as a positive control in molecular docking and MDS analyses. RESULTS We report here the identification of a few novel intracellular tripeptides, namely Arg-His- Trp, (PubChem CID-145453842), Pro-Ile-Glu, (PubChem CID-145457492), Cys-Gln-Gln, (PubChem CID-71471965), Glu-Glu-Lys, (PubChem CID-11441068) and Gly-Cys-Leu (PubChem CID-145455600) of FBS origin in MCF-7 cells. Molecular docking and MDS analyses revealed that among these molecules, the tripeptide Arg-His-Trp shows a favorable binding affinity with β2AR (-9.8 Kcal/mol). The agonistic effect of Arg-His-Trp is significant and comparable with that of a known agonist of β2AR, isoproterenol. CONCLUSION In conclusion, we identified a unique Arg-His-Trp tripeptide of FBS origin in MCF-7 cells by employing a novel approach. This unique tripeptide Arg-His-Trp is suggested to be a potential agonist of β2AR and it may have applications in the context of various human diseases like bronchial asthma and chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Hritik Chandore
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| | - Ajay Kumar Raj
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| | - Kiran Bharat Lokhande
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| | - Krishna Venkateswara Swamy
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| | - Jayanta Kumar Pal
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| |
Collapse
|
32
|
Gensbittel V, Kräter M, Harlepp S, Busnelli I, Guck J, Goetz JG. Mechanical Adaptability of Tumor Cells in Metastasis. Dev Cell 2020; 56:164-179. [PMID: 33238151 DOI: 10.1016/j.devcel.2020.10.011] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/18/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
The most dangerous aspect of cancer lies in metastatic progression. Tumor cells will successfully form life-threatening metastases when they undergo sequential steps along a journey from the primary tumor to distant organs. From a biomechanics standpoint, growth, invasion, intravasation, circulation, arrest/adhesion, and extravasation of tumor cells demand particular cell-mechanical properties in order to survive and complete the metastatic cascade. With metastatic cells usually being softer than their non-malignant counterparts, high deformability for both the cell and its nucleus is thought to offer a significant advantage for metastatic potential. However, it is still unclear whether there is a finely tuned but fixed mechanical state that accommodates all mechanical features required for survival throughout the cascade or whether tumor cells need to dynamically refine their properties and intracellular components at each new step encountered. Here, we review the various mechanical requirements successful cancer cells might need to fulfill along their journey and speculate on the possibility that they dynamically adapt their properties accordingly. The mechanical signature of a successful cancer cell might actually be its ability to adapt to the successive microenvironmental constraints along the different steps of the journey.
Collapse
Affiliation(s)
- Valentin Gensbittel
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Sébastien Harlepp
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Ignacio Busnelli
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
33
|
Sakakitani S, Podyma-Inoue KA, Takayama R, Takahashi K, Ishigami-Yuasa M, Kagechika H, Harada H, Watabe T. Activation of β2-adrenergic receptor signals suppresses mesenchymal phenotypes of oral squamous cell carcinoma cells. Cancer Sci 2020; 112:155-167. [PMID: 33007125 PMCID: PMC7780019 DOI: 10.1111/cas.14670] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022] Open
Abstract
Metastasis is a primary reason related to the mortality of oral squamous cell carcinoma (OSCC) patients. A program called epithelial-mesenchymal transition (EMT) has been shown to play a critical role in promoting metastasis in epithelium-derived carcinoma. During EMT, epithelial cancer cells acquire motile mesenchymal phenotypes and detach from primary tumors. Recent lines of evidence have suggested that EMT confers cancer cells with tumor-initiating ability. Therefore, selective targeting of EMT would lead to the development of effective therapeutic agents. In this study, using a chemical biology approach, we identified isoxsuprine, a β2-adrenergic receptor (β2-AR) agonist as a low-molecular-weight compound that interferes with the acquisition of mesenchymal phenotypes of oral cancer cells. Treatment of multiple types of oral cancer cells with isoxsuprine led to the downregulation of mesenchymal cell markers that was accompanied by reduced cell motility. Similar inhibitory effects were also observed for isoprenaline, a non-selective β-adrenergic receptor (β-AR) agonist. In addition, inhibition of cell migration upon treatment with isoxsuprine was reverted by a non-selective β-AR antagonist, propranolol, and the CRISPR/Cas9 system-mediated deletion of the β2-AR gene, suggesting that the effects exerted by isoxsuprine involved signals mediated by β2-AR. In addition, in a subcutaneous xenograft model of oral cancer cells, the administration of isoxsuprine effectively suppressed primary tumor growth, suggesting β2-AR signals to be a promising cancer therapeutic target for treatment of OSCC.
Collapse
Affiliation(s)
- Shintaro Sakakitani
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Katarzyna A Podyma-Inoue
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Rina Takayama
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazuki Takahashi
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Mari Ishigami-Yuasa
- Department of Organic and Medicinal Chemistry, Chemical Biology Screening Center, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroyuki Kagechika
- Department of Organic and Medicinal Chemistry, Chemical Biology Screening Center, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
34
|
Mravec B, Horvathova L, Hunakova L. Neurobiology of Cancer: the Role of β-Adrenergic Receptor Signaling in Various Tumor Environments. Int J Mol Sci 2020; 21:ijms21217958. [PMID: 33114769 PMCID: PMC7662752 DOI: 10.3390/ijms21217958] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
The development and progression of cancer depends on both tumor micro- and macroenvironments. In addition, psychosocial and spiritual “environments” might also affect cancer. It has been found that the nervous system, via neural and humoral pathways, significantly modulates processes related to cancer at the level of the tumor micro- and macroenvironments. The nervous system also mediates the effects of psychosocial and noetic factors on cancer. Importantly, data accumulated in the last two decades have clearly shown that effects of the nervous system on cancer initiation, progression, and the development of metastases are mediated by the sympathoadrenal system mainly via β-adrenergic receptor signaling. Here, we provide a new complex view of the role of β-adrenergic receptor signaling within the tumor micro- and macroenvironments as well as in mediating the effects of the psychosocial and spiritual environments. In addition, we describe potential preventive and therapeutic implications.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, 814 39 Bratislava, Slovakia;
- Correspondence: ; Tel.: +421-(2)-59357527; Fax: +421-(2)-59357601
| | - Lubica Horvathova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, 814 39 Bratislava, Slovakia;
| | - Luba Hunakova
- Institute of Microbiology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia;
| |
Collapse
|
35
|
Yokota T, McCourt J, Ma F, Ren S, Li S, Kim TH, Kurmangaliyev YZ, Nasiri R, Ahadian S, Nguyen T, Tan XHM, Zhou Y, Wu R, Rodriguez A, Cohn W, Wang Y, Whitelegge J, Ryazantsev S, Khademhosseini A, Teitell MA, Chiou PY, Birk DE, Rowat AC, Crosbie RH, Pellegrini M, Seldin M, Lusis AJ, Deb A. Type V Collagen in Scar Tissue Regulates the Size of Scar after Heart Injury. Cell 2020; 182:545-562.e23. [PMID: 32621799 PMCID: PMC7415659 DOI: 10.1016/j.cell.2020.06.030] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/17/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022]
Abstract
Scar tissue size following myocardial infarction is an independent predictor of cardiovascular outcomes, yet little is known about factors regulating scar size. We demonstrate that collagen V, a minor constituent of heart scars, regulates the size of heart scars after ischemic injury. Depletion of collagen V led to a paradoxical increase in post-infarction scar size with worsening of heart function. A systems genetics approach across 100 in-bred strains of mice demonstrated that collagen V is a critical driver of postinjury heart function. We show that collagen V deficiency alters the mechanical properties of scar tissue, and altered reciprocal feedback between matrix and cells induces expression of mechanosensitive integrins that drive fibroblast activation and increase scar size. Cilengitide, an inhibitor of specific integrins, rescues the phenotype of increased post-injury scarring in collagen-V-deficient mice. These observations demonstrate that collagen V regulates scar size in an integrin-dependent manner.
Collapse
Affiliation(s)
- Tomohiro Yokota
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Jackie McCourt
- Department of Integrative Biology and Physiology, University of California, CA 90095, USA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Shuxun Ren
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Tae-Hyung Kim
- Department of Integrative Biology and Physiology, University of California, CA 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Rohollah Nasiri
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Samad Ahadian
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Thang Nguyen
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA
| | - Xing Haw Marvin Tan
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
| | - Yonggang Zhou
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Rimao Wu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Abraham Rodriguez
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Whitaker Cohn
- Passarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience and Behaviour, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Yibin Wang
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Julian Whitelegge
- Passarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience and Behaviour, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Sergey Ryazantsev
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Ali Khademhosseini
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA; Department of Chemical Engineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Michael A Teitell
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Pei-Yu Chiou
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
| | - David E Birk
- University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Marcus Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, CA 92697, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Genetics, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Nguyen AK, Kilian KA. Physicochemical Tools for Visualizing and Quantifying Cell-Generated Forces. ACS Chem Biol 2020; 15:1731-1746. [PMID: 32530602 DOI: 10.1021/acschembio.0c00304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To discern how mechanical forces coordinate biological outcomes, methods that map cell-generated forces in a spatiotemporal manner, and at cellular length scales, are critical. In their native environment, whether it be within compact multicellular three-dimensional structures or sparsely populated fibrillar networks of the extracellular matrix, cells are constantly exposed to a slew of physical forces acting on them from all directions. At the same time, cells exert highly localized forces of their own on their surroundings and on neighboring cells. Together, the generation and transmission of these forces can control diverse cellular activities and behavior as well as influence cell fate decisions. To thoroughly understand these processes, we must first be able to characterize and measure such forces. However, our experimental needs and technical capabilities are in discord-while it is apparent that we should study cell-generated forces within more biologically relevant 3D environments, this goal remains challenging because of caveats associated with complex "sensing-transduction-readout" modalities. In this Review, we will discuss the latest techniques for measuring cell-generated forces. We will highlight recent advances in traction force microscopy and examine new alternative approaches for quantifying cell-generated forces, both of individual cells and within 3D tissues. Finally, we will explore the future direction of novel cellular force-sensing tools in the context of mechanobiology and next-generation biomaterials design.
Collapse
Affiliation(s)
- Ashley K. Nguyen
- School of Chemistry, School of Materials Science and Engineering, Australian Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Kristopher A. Kilian
- School of Chemistry, School of Materials Science and Engineering, Australian Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
37
|
Kwon S, Yang W, Moon D, Kim KS. Comparison of Cancer Cell Elasticity by Cell Type. J Cancer 2020; 11:5403-5412. [PMID: 32742487 PMCID: PMC7391204 DOI: 10.7150/jca.45897] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/24/2020] [Indexed: 12/27/2022] Open
Abstract
Lower cellular elasticity is a distinguishing feature of cancer cells compared with normal cells. To determine whether cellular elasticity differs based on cancer cell type, cells were selected from three different cancer types including breast, cervix, and lung. For each cancer type, one counterpart normal cell and three types of cancer cells were selected, and their elasticity was measured using atomic force microscopy (AFM). The elasticity of normal cells was in the order of MCF10A > WI-38 ≥ Ect1/E6E7 which corresponds to the counterpart normal breast, lung, and cervical cancer cells, respectively. All cancer cells exhibited lower elasticity than their counterpart normal cells. Compared with the counterpart normal cells, the difference in cellular elasticity was the greatest in cervical cancer cells, followed by lung and breast cancer cells. This result indicates lower elasticity is a unique property of cancer cells; however, the reduction in elasticity may depend on the histological origin of the cells. The F-actin cytoskeleton of cancer cells was different in structure and content from normal cells. The F-actin is mainly distributed at the periphery of cancer cells and its content was mostly lower than that seen in normal cells.
Collapse
Affiliation(s)
- Sangwoo Kwon
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 130-710, Republic of Korea
| | - Woochul Yang
- Department of Physics, Dongguk University, Seoul 04620, Republic of Korea
| | - Donggerami Moon
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 130-710, Republic of Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul 130-710, Republic of Korea
| |
Collapse
|
38
|
Zhang C, Liao X, Ma Z, Liu S, Fang F, Mai H. Overexpression of β-Adrenergic Receptors and the Suppressive Effect of β 2-Adrenergic Receptor Blockade in Oral Squamous Cell Carcinoma. J Oral Maxillofac Surg 2020; 78:1871.e1-1871.e23. [PMID: 32640209 DOI: 10.1016/j.joms.2020.05.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE The purpose of this project was to investigate the expression of β-adrenergic receptors in oral squamous cell carcinoma (OSCC) and the tumor suppressive activity of β2-adrenergic receptor (β2-AR) blockade. MATERIALS AND METHODS Samples of 15 normal oral mucosal epithelial tissues, 60 surgically resected OSCC tissues, and 60 adjacent para-carcinoma tissues were collected. The expression of β1-adrenergic receptor and β2-AR was detected by real-time quantitative polymerase chain reaction and the Western blot test. SCC9 and Cal27 cell lines and primary OSCC cells also were included and treated with ICI-118,551 (MedChemExpress, Monmouth Junction, NJ), a selective β2-AR blocker. In addition, the Cal27 cell line was treated with propranolol (a nonselective β-adrenergic receptor blocker) to verify the suppressive effect of β2-AR blockade. For in vivo assays, Cal27 cells were subcutaneously injected in the tongue flank of nude mice. ICI-118,551 was orally administered to the mice in the treatment group daily. High-throughput sequencing was used to screen for changes in gene expression. RESULTS Real-time quantitative polymerase chain reaction and the Western blot test both showed that β1-adrenergic receptor and β2-AR were overexpressed in OSCC tissues and cells. A relationship was found between β2-AR and a more advanced clinical stage, as well as preoperative lymphatic metastasis. After treatment with ICI-118,551 or propranolol, the capacities for proliferation, invasion, and metastasis of OSCC cells were significantly inhibited. Tumor size was significantly different between the ICI-118,551 and control groups. The survival time in the ICI-118,551 group also was prolonged significantly. Moreover, high-throughput sequencing identified 19 affected signaling pathways, including mitogen-activated protein kinase and PI3K-Akt. We confirmed a significant change to the expression of several genes closely related to the progression of cancer. CONCLUSION This study showed that β2-AR is related to a more advanced clinical stage and preoperative lymphatic metastasis. Additionally, a β2-AR blocker has a significant suppressive effect in OSCC.
Collapse
Affiliation(s)
- Chong Zhang
- Resident, Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment; and Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Xianxiang Liao
- Resident, Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment; and Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Zhen Ma
- Resident, Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment; and Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Shiqi Liu
- Resident, Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment; and Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Fang Fang
- Resident, Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment; and Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China
| | - Huaming Mai
- Professor, Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Guangxi Medical University; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment; and Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, China.
| |
Collapse
|
39
|
Hymel SJ, Lan H, Khismatullin DB. Elongation Index as a Sensitive Measure of Cell Deformation in High-Throughput Microfluidic Systems. Biophys J 2020; 119:493-501. [PMID: 32697978 DOI: 10.1016/j.bpj.2020.06.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/25/2020] [Accepted: 06/24/2020] [Indexed: 01/10/2023] Open
Abstract
One of the promising approaches for high-throughput screening of cell mechanotype is microfluidic deformability cytometry (mDC), in which the apparent deformation index (DI) of the cells stretched by extensional flow at the stagnation point of a cross-slot microchannel is measured. The DI is subject to substantial measurement errors due to cell offset from the flow centerline and velocity fluctuations in inlet channels, leading to artificial widening of DI versus cell size plots. Here, we simulated an mDC experiment using a custom computational algorithm for viscoelastic cell migration. Cell motion and deformation in a cross-slot channel was modeled for fixed or randomized values of cellular mechanical properties (diameter, shear elasticity, cortical tension) and initial cell placement, with or without sinusoidal fluctuations between the inlet velocities. Our numerical simulation indicates that mDC loses sensitivity to changes in shear elasticity when the offset distance exceeds 5 μm, and just 1% velocity fluctuation causes an 11.7% drop in the DI. The obtained relationships between the cell diameter, shear elasticity, and offset distance were used to establish a new measure of cell deformation, referred to as the "elongation index" (EI). In the randomized study, the EI scatter plots were visibly separated for the low- and high-elasticity populations of cells, with a mean of 300 and 3500 Pa, whereas the standard DI output was unable to distinguish between these two groups of cells. The successful suppression of the offset artifacts with a narrower data distribution was shown for the EI output of MCF-7 cells.
Collapse
Affiliation(s)
- Scott J Hymel
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Hongzhi Lan
- Department of Pediatrics, Stanford University, Stanford, California
| | - Damir B Khismatullin
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana.
| |
Collapse
|
40
|
Mravec B, Tibensky M, Horvathova L. Stress and cancer. Part II: Therapeutic implications for oncology. J Neuroimmunol 2020; 346:577312. [PMID: 32652364 DOI: 10.1016/j.jneuroim.2020.577312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
Accumulated evidence has confirmed the ability of stress to promote the induction and progression of cancer (for review see Stress and cancer. Part I: Mechanisms mediating the effect of stressors on cancer). In support of this, data from clinical trials utilizing approaches that reduce stress-related signaling have shown prolonged survival of cancer patients. Therefore, the question has arisen as to how we can utilize this knowledge in the daily treatment of cancer patients. The main aim of this review is to critically analyze data from studies utilizing psychotherapy or treatment by β-blockers on the survival of cancer patients. Because these approaches, especially treatment by β-blockers, have been routinely used in clinical practice for decades in the treatment of non-cancer patients, their wider introduction into oncology might be realized in the near future.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia; Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Miroslav Tibensky
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia
| | - Lubica Horvathova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
41
|
Mravec B, Tibensky M, Horvathova L. Stress and cancer. Part I: Mechanisms mediating the effect of stressors on cancer. J Neuroimmunol 2020; 346:577311. [PMID: 32652365 DOI: 10.1016/j.jneuroim.2020.577311] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
Observations indicating a link between psychosocial stress and cancer can be traced back almost 2 millennia. However, the pathways and mechanisms interconnecting them has only been elucidated in more detail since the end of the 20th century. Importantly, recently accumulated evidences have confirmed the ability of stress to promote the induction and progression of cancer. The main aim of this review is to describe the pathways and mechanisms mediating the stimulatory effects of the neuroendocrine stress response on the induction of cancer, potentiation of cancer growth, and the development of metastases.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia; Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Miroslav Tibensky
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia
| | - Lubica Horvathova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
42
|
β2-Adrenergic Signalling Promotes Cell Migration by Upregulating Expression of the Metastasis-Associated Molecule LYPD3. BIOLOGY 2020; 9:biology9020039. [PMID: 32098331 PMCID: PMC7168268 DOI: 10.3390/biology9020039] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/13/2020] [Accepted: 02/20/2020] [Indexed: 12/24/2022]
Abstract
Metastasis is associated with poor prognosis in breast cancer. Although some studies suggest beta-blockers increase survival by delaying metastasis, others have been discordant. This study provides both insights into the anomalous findings and identifies potential biomarkers that may be treatment targets. Cell line models of basal-type and oestrogen receptor-positive breast cancer were profiled for basal levels of adrenoceptor gene/protein expression, and β2-adrenoceptor mediated cell behaviour including migration, invasion, adhesion, and survival in response to adrenoceptor agonist/antagonist treatment. Protein profiling and histology identified biomarkers and drug targets. Baseline levels of adrenoceptor gene expression are higher in basal-type rather than oestrogen receptor-positive cancer cells. Norepinephrine (NE) treatment increased invasive capacity in all cell lines but did not increase proliferation/survival. Protein profiling revealed the upregulation of the pro-metastatic gene Ly6/PLAUR Domain-Containing Protein 3 (LYPD3) in norepinephrine-treated MDA-MB-468 cells. Histology confirmed selective LYPD3 expression in primary and metastatic breast tumour samples. These findings demonstrate that basal-type cancer cells show a more aggressive adrenoceptor-β2-activated phenotype in the resting and stimulated state, which is attenuated by adrenoceptor-β2 inhibition. This study also highlights the first association between ADRβ2 signalling and LYPD3; its knockdown significantly reduced the basal and norepinephrine-induced activity of MCF-7 cells in vitro. The regulation of ADRβ2 signalling by LYPD3 and its metastasis promoting activities, reveal LYPD3 as a promising therapeutic target in the treatment of breast and other cancers.
Collapse
|
43
|
Follain G, Herrmann D, Harlepp S, Hyenne V, Osmani N, Warren SC, Timpson P, Goetz JG. Fluids and their mechanics in tumour transit: shaping metastasis. Nat Rev Cancer 2020; 20:107-124. [PMID: 31780785 DOI: 10.1038/s41568-019-0221-x] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Metastasis is a dynamic succession of events involving the dissemination of tumour cells to distant sites within the body, ultimately reducing the survival of patients with cancer. To colonize distant organs and, therefore, systemically disseminate within the organism, cancer cells and associated factors exploit several bodily fluid systems, which provide a natural transportation route. Indeed, the flow mechanics of the blood and lymphatic circulatory systems can be co-opted to improve the efficiency of cancer cell transit from the primary tumour, extravasation and metastatic seeding. Flow rates, vessel size and shear stress can all influence the survival of cancer cells in the circulation and control organotropic seeding patterns. Thus, in addition to using these fluids as a means to travel throughout the body, cancer cells exploit the underlying physical forces within these fluids to successfully seed distant metastases. In this Review, we describe how circulating tumour cells and tumour-associated factors leverage bodily fluids, their underlying forces and imposed stresses during metastasis. As the contribution of bodily fluids and their mechanics raises interesting questions about the biology of the metastatic cascade, an improved understanding of this process might provide a new avenue for targeting cancer cells in transit.
Collapse
Affiliation(s)
- Gautier Follain
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - David Herrmann
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Sébastien Harlepp
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Vincent Hyenne
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- CNRS SNC 505, Strasbourg, France
| | - Naël Osmani
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Sean C Warren
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
44
|
Nguyen AV, Trompetto B, Tan XHM, Scott MB, Hu KHH, Deeds E, Butte MJ, Chiou PY, Rowat AC. Differential Contributions of Actin and Myosin to the Physical Phenotypes and Invasion of Pancreatic Cancer Cells. Cell Mol Bioeng 2020; 13:27-44. [PMID: 32030106 PMCID: PMC6981337 DOI: 10.1007/s12195-019-00603-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Metastasis is a fundamentally physical process in which cells deform through narrow gaps and generate forces to invade surrounding tissues. While it is commonly thought that increased cell deformability is an advantage for invading cells, we previously found that more invasive pancreatic ductal adenocarcinoma (PDAC) cells are stiffer than less invasive PDAC cells. Here we investigate potential mechanisms of the simultaneous increase in PDAC cell stiffness and invasion, focusing on the contributions of myosin II, Arp2/3, and formins. METHOD We measure cell invasion using a 3D scratch wound invasion assay and cell stiffness using atomic force microscopy (AFM). To determine the effects of actin- and myosin-mediated force generation on cell stiffness and invasion, we treat cells with pharmacologic inhibitors of myosin II (blebbistatin), Arp2/3 (CK-666), and formins (SMIFH2). RESULTS We find that the activity of myosin II, Arp2/3, and formins all contribute to the stiffness of PDAC cells. Interestingly, we find that the invasion of PDAC cell lines is differentially affected when the activity of myosin II, Arp2/3, or formins is inhibited, suggesting that despite having similar tissue origins, different PDAC cell lines may rely on different mechanisms for invasion. CONCLUSIONS These findings deepen our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incite further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit.
Collapse
Affiliation(s)
- Angelyn V. Nguyen
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | - Brittany Trompetto
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | | | - Michael B. Scott
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
- Present Address: Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, USA
- Department of Biomedical Engineering, Northwestern McCormick School of Engineering, Evanston, USA
| | | | - Eric Deeds
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, USA
| | - Manish J. Butte
- Department of Pediatrics, University of California, Los Angeles, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, USA
| | - Pei Yu Chiou
- Department of Bioengineering, University of California, Los Angeles, USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
| | - Amy C. Rowat
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, USA
| |
Collapse
|
45
|
Chen M, Zeng J, Ruan W, Zhang Z, Wang Y, Xie S, Wang Z, Yang H. Examination of the relationship between viscoelastic properties and the invasion of ovarian cancer cells by atomic force microscopy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:568-582. [PMID: 32318318 PMCID: PMC7155897 DOI: 10.3762/bjnano.11.45] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 03/04/2020] [Indexed: 05/17/2023]
Abstract
The mechanical properties of cells could serve as an indicator for disease progression and early cancer diagnosis. This study utilized atomic force microscopy (AFM) to measure the viscoelastic properties of ovarian cancer cells and then examined the association with the invasion of ovarian cancer at the level of living single cells. Elasticity and viscosity of the ovarian cancer cells OVCAR-3 and HO-8910 are significantly lower than those of the human ovarian surface epithelial cell (HOSEpiC) control. Further examination found a dramatic increase of migration/invasion and an obvious decease of microfilament density in OVCAR-3 and HO-8910 cells. Also, there was a significant relationship between viscoelastic and biological properties among these cells. In addition, the elasticity was significantly increased in OVCAR-3 and HO-8910 cells after the treatment with the anticancer compound echinomycin (Ech), while no obvious change was found in HOSEpiC cells after Ech treatment. Interestingly, Ech seemed to have no effect on the viscosity of the cells. Ech significantly inhibited the migration/invasion and significantly increased the microfilament density in OVCAR-3 and HO-8910 cells, which was significantly related with the elasticity of the cells. An increase of elasticity and a decrease of invasion were found in OVCAR-3 and HO-8910 cells after Ech treatment. Together, this study clearly demonstrated the association of viscoelastic properties with the invasion of ovarian cancer cells and shed a light on the biomechanical changes for early diagnosis of tumor transformation and progression at single-cell level.
Collapse
Affiliation(s)
- Mengdan Chen
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Jinshu Zeng
- Department of Ultrasound Medical, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Weiwei Ruan
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Zhenghong Zhang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Yuhua Wang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Shusen Xie
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Zhengchao Wang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Hongqin Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| |
Collapse
|
46
|
Surcel A, Schiffhauer ES, Thomas DG, Zhu Q, DiNapoli KT, Herbig M, Otto O, West-Foyle H, Jacobi A, Kräter M, Plak K, Guck J, Jaffee EM, Iglesias PA, Anders RA, Robinson DN. Targeting Mechanoresponsive Proteins in Pancreatic Cancer: 4-Hydroxyacetophenone Blocks Dissemination and Invasion by Activating MYH14. Cancer Res 2019; 79:4665-4678. [PMID: 31358530 PMCID: PMC6744980 DOI: 10.1158/0008-5472.can-18-3131] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 06/11/2019] [Accepted: 07/22/2019] [Indexed: 12/16/2022]
Abstract
Metastasis is complex, involving multiple genetic, epigenetic, biochemical, and physical changes in the cancer cell and its microenvironment. Cells with metastatic potential are often characterized by altered cellular contractility and deformability, lending them the flexibility to disseminate and navigate through different microenvironments. We demonstrate that mechanoresponsiveness is a hallmark of pancreatic cancer cells. Key mechanoresponsive proteins, those that accumulate in response to mechanical stress, specifically nonmuscle myosin IIA (MYH9) and IIC (MYH14), α-actinin 4, and filamin B, were highly expressed in pancreatic cancer as compared with healthy ductal epithelia. Their less responsive sister paralogs-myosin IIB (MYH10), α-actinin 1, and filamin A-had lower expression differential or disappeared with cancer progression. We demonstrate that proteins whose cellular contributions are often overlooked because of their low abundance can have profound impact on cell architecture, behavior, and mechanics. Here, the low abundant protein MYH14 promoted metastatic behavior and could be exploited with 4-hydroxyacetophenone (4-HAP), which increased MYH14 assembly, stiffening cells. As a result, 4-HAP decreased dissemination, induced cortical actin belts in spheroids, and slowed retrograde actin flow. 4-HAP also reduced liver metastases in human pancreatic cancer-bearing nude mice. Thus, increasing MYH14 assembly overwhelms the ability of cells to polarize and invade, suggesting targeting the mechanoresponsive proteins of the actin cytoskeleton as a new strategy to improve the survival of patients with pancreatic cancer. SIGNIFICANCE: This study demonstrates that mechanoresponsive proteins become upregulated with pancreatic cancer progression and that this system of proteins can be pharmacologically targeted to inhibit the metastatic potential of pancreatic cancer cells.
Collapse
Affiliation(s)
- Alexandra Surcel
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Eric S Schiffhauer
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dustin G Thomas
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qingfeng Zhu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kathleen T DiNapoli
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Electrical and Computer Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland
| | - Maik Herbig
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Oliver Otto
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Hoku West-Foyle
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Angela Jacobi
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Martin Kräter
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Katarzyna Plak
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins, The Skip Viragh Pancreatic Cancer Center, and the Bloomberg Kimmel Institute, Johns Hopkins University, Baltimore, Maryland
| | - Pablo A Iglesias
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Electrical and Computer Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland
| | - Robert A Anders
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
47
|
Sloan EK, Walker AK. Elucidating the mechanisms of psychosocial influences on cancer using preclinical in vivo models. Curr Opin Behav Sci 2019. [DOI: 10.1016/j.cobeha.2019.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
48
|
Gill NK, Ly C, Kim PH, Saunders CA, Fong LG, Young SG, Luxton GWG, Rowat AC. DYT1 Dystonia Patient-Derived Fibroblasts Have Increased Deformability and Susceptibility to Damage by Mechanical Forces. Front Cell Dev Biol 2019; 7:103. [PMID: 31294022 PMCID: PMC6606767 DOI: 10.3389/fcell.2019.00103] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/27/2019] [Indexed: 12/24/2022] Open
Abstract
DYT1 dystonia is a neurological movement disorder that is caused by a loss-of-function mutation in the DYT1/TOR1A gene, which encodes torsinA, a conserved luminal ATPases-associated with various cellular activities (AAA+) protein. TorsinA is required for the assembly of functional linker of nucleoskeleton and cytoskeleton (LINC) complexes, and consequently the mechanical integration of the nucleus and the cytoskeleton. Despite the potential implications of altered mechanobiology in dystonia pathogenesis, the role of torsinA in regulating cellular mechanical phenotype, or mechanotype, in DYT1 dystonia remains unknown. Here, we define the deformability of mouse fibroblasts lacking functional torsinA as well as human fibroblasts isolated from DYT1 dystonia patients. We find that the deletion of torsinA or the expression of torsinA containing the DYT1 dystonia-causing ΔE302/303 (ΔE) mutation results in more deformable cells. We observe a similar increased deformability of mouse fibroblasts that lack lamina-associated polypeptide 1 (LAP1), which interacts with and stimulates the ATPase activity of torsinA in vitro, as well as with the absence of the LINC complex proteins, Sad1/UNC-84 1 (SUN1) and SUN2, lamin A/C, or lamin B1. Consistent with these findings, we also determine that DYT1 dystonia patient-derived fibroblasts are more compliant than fibroblasts isolated from unafflicted individuals. DYT1 dystonia patient-derived fibroblasts also exhibit increased nuclear strain and decreased viability following mechanical stretch. Taken together, our results establish the foundation for future mechanistic studies of the role of cellular mechanotype and LINC-dependent nuclear-cytoskeletal coupling in regulating cell survival following exposure to mechanical stresses.
Collapse
Affiliation(s)
- Navjot Kaur Gill
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Chau Ly
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Paul H Kim
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cosmo A Saunders
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, United States
| | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, United States.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - G W Gant Luxton
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, United States.,Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
49
|
Lee A, Fraser SP, Djamgoz MBA. Propranolol inhibits neonatal Nav1.5 activity and invasiveness of MDA-MB-231 breast cancer cells: Effects of combination with ranolazine. J Cell Physiol 2019; 234:23066-23081. [PMID: 31222761 DOI: 10.1002/jcp.28868] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/11/2019] [Accepted: 04/17/2019] [Indexed: 01/08/2023]
Abstract
The MDA-MB-231 cell line was used as a model of triple negative breast cancer to investigate the interaction of β-adrenergic receptor (β-AR) and voltage-gated sodium channel (VGSC). There was significant (86%) overlap in their expression. Short-term (acute) application of the β-AR antagonist propranolol (25 μM) led to reduction of peak current and quickening of current inactivation (the latter occurred only in 5% fetal bovine serum). Long-term (48 hr) incubation with propranolol (25 μM) resulted in several changes in VGSC characteristics: shifts in (a) current-voltage relationship and (b) steady-state inactivation, both to more negative potentials and (c) the slowing of recovery from inactivation. We then investigated the effects of propranolol and ranolazine, a blocker of VGSC activity, alone and in combination, on lateral motility and Matrigel invasion. These assays were carried out under hypoxic conditions more representative of tumor progression. Propranolol (2.5 and 25 μM) and ranolazine (5 μM), and their combination inhibited lateral motility. Also, propranolol (25 μM) and ranolazine (5 μM), and their combination inhibited invasion. However, no synergy was observed in the pharmacological combinations for both assays. Propranolol also significantly decreased total neonatal Nav1.5 protein expression, the predominant VGSC subtype expressed in these cells. We conclude (a) that β-AR and VGSC are functionally coupled in MDA-MB-231 cells; (b) that propranolol has direct blocking action on the VGSC; (c) that the action of propranolol is modulated by serum; and (d) that the antimetastatic cellular effects of propranolol and ranolazine are not additive.
Collapse
Affiliation(s)
- Alice Lee
- Faculty of Medicine, Imperial College London, South Kensington Campus, London, UK
| | - Scott P Fraser
- Department of Life Sciences, Faculty of Natural Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London, UK
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Faculty of Natural Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London, UK
| |
Collapse
|
50
|
Hanns P, Paczulla AM, Medinger M, Konantz M, Lengerke C. Stress and catecholamines modulate the bone marrow microenvironment to promote tumorigenesis. Cell Stress 2019; 3:221-235. [PMID: 31338489 PMCID: PMC6612892 DOI: 10.15698/cst2019.07.192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
High vascularization and locally secreted factors make the bone marrow (BM) microenvironment particularly hospitable for tumor cells and bones to a preferred metastatic site for disseminated cancer cells of different origins. Cancer cell homing and proliferation in the BM are amongst other regulated by complex interactions with BM niche cells (e.g. osteoblasts, endothelial cells and mesenchymal stromal cells (MSCs)), resident hematopoietic stem and progenitor cells (HSPCs) and pro-angiogenic cytokines leading to enhanced BM microvessel densities during malignant progression. Stress and catecholamine neurotransmitters released in response to activation of the sympathetic nervous system (SNS) reportedly modulate various BM cells and may thereby influence cancer progression. Here we review the role of catecholamines during tumorigenesis with particular focus on pro-tumorigenic effects mediated by the BM niche.
Collapse
Affiliation(s)
- Pauline Hanns
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Anna M Paczulla
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Michael Medinger
- Division of Clinical Hematology, University Hospital Basel, Basel, Switzerland
| | - Martina Konantz
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Claudia Lengerke
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland.,Division of Clinical Hematology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|