1
|
Yu F, Zhang G, Sun J, Zhao Y, Qi Y, Han X, Ai C, Sun W, Duan J, Yu D. Nanotension Relief Agent Enhances Tissue Penetration by Reducing Solid Stress in Pancreatic Ductal Adenocarcinoma via Rho/ROCK Pathway Inhibition. Biomater Res 2025; 29:0173. [PMID: 40207257 PMCID: PMC11979343 DOI: 10.34133/bmr.0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
The formidable contractile tension exerted by cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinoma (PDAC) tissue is crucial for maintaining high tissue solid stress (TSS), which impedes the delivery and penetration of chemotherapeutic drugs. To address this obstacle, we constructed a pH-responsive nanotension relief agent (FS@MMS), in which fasudil (FS) was ingeniously conjugated to mesoporous silica encapsulated with magnetic iron oxide (MMS). The nanotension relief agent was demonstrated to inhibit the synthesis of phosphorylated myosin light chain by blocking the Rho/Rho-associated serine/threonine kinase (ROCK) pathway, triggering the swift transformation of high-tension CAFs into low-tension CAFs in PDAC tissue, which relieves TSS and enhances drug penetration in Panc02/NIH-3T3 multicellular tumor spheroids. When the nanotension relief agent was further loaded with the chemotherapeutic drug gemcitabine (GEM), as FS@MMS-GEM, the enhanced permeation of GEM progressively killed tumor cells and amplified their TSS-relief properties, thereby maximizing the anticancer efficacy of chemotherapeutic agents in Panc02/NIH-3T3 coplanted model mice. The magnetic resonance imaging results revealed that the synergistic effect substantially improved drug delivery and penetration efficiency. The developed approach holds great potential for improving chemotherapy efficacy in PDAC and provides a novel therapeutic approach for the treatment of related stroma-rich tumors.
Collapse
Affiliation(s)
- Feiran Yu
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Gaorui Zhang
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jintang Sun
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yuxuan Zhao
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yafei Qi
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiaoyu Han
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Chen Ai
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Weikai Sun
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jiazhi Duan
- Institute for Advanced Interdisciplinary Research,
University of Jinan, Jinan 250022, China
| | - Dexin Yu
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
| |
Collapse
|
2
|
Guan X, Shen Y, Zhao C, Li X, Li X, Lu D, Wang L, Liu L, Wu S, Huang B, Guo L, Xu H. Cascade-Responsive Nanoprodrug Disrupts Immune-Fibroblast Communications for Potentiated Cancer Mechanoimmunotherapy. Adv Healthc Mater 2025:e2500176. [PMID: 40079115 DOI: 10.1002/adhm.202500176] [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: 01/10/2025] [Revised: 03/02/2025] [Indexed: 03/14/2025]
Abstract
The abnormal tumor mechanical microenvironment due to specific cancer-associated fibroblasts (CAFs) subset and low tumor immunogenicity caused by inefficient conversion of active chemotherapeutic agents are two key obstacles that impede patients with desmoplastic tumors from achieving stable and complete immune responses. Herein, it is demonstrated that FAP-α+CAFs-induced stromal stiffness accelerated tumor progression by precluding cytotoxic T lymphocytes. Subsequently, a cascade-responsive nanoprodrug capable of re-educating FAP-α+CAFs and amplifying tumor immunogenicity for potentiated cancer mechanoimmunotherapy is ingeniously designed. Benefiting from the active targeted release of angiotensin II receptor antagonist (losartan) guided by FAP-α cleavable peptide and the efficient conversion of topoisomerase I inhibitor (7-Ethyl-10-hydroxycamptothecin) prodrug under high glutathione/esterase within tumor cells, this regimen created an immune-activated landscape that retarded primary tumor growth and counteracted resistance to immune checkpoint inhibitor in mice with triple-negative breast cancer. This nanoprodrug-assisted mechanoimmunotherapy can serve as a universal strategy for conferring efficient tumoricidal immunity in "immune excluded" desmoplastic tumor interventions.
Collapse
Affiliation(s)
- Xin Guan
- Department of Ultrasound, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, P. R. China
| | - Yuting Shen
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chongke Zhao
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xiao Li
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xiaolong Li
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Dan Lu
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Lifan Wang
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Linna Liu
- Department of Ultrasound, Institiute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Shengbo Wu
- Department of Ultrasound, Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Bin Huang
- Department of Ultrasound, Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Lehang Guo
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, P. R. China
| |
Collapse
|
3
|
Zhang Y, Fu Q, Sun W, Yue Q, He P, Niu D, Zhang M. Mechanical forces in the tumor microenvironment: roles, pathways, and therapeutic approaches. J Transl Med 2025; 23:313. [PMID: 40075523 PMCID: PMC11899831 DOI: 10.1186/s12967-025-06306-8] [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: 11/26/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
Tumors often exhibit greater stiffness compared to normal tissues, primarily due to increased deposition within the tumor stroma. Collagen, proteoglycans, laminin, and fibronectin are key components of the extracellular matrix (ECM), interacting to facilitate ECM assembly. Enhanced fiber density and cross-linking within the ECM result in elevated matrix stiffness and interstitial fluid pressure, subjecting tumors to significant physical stress during growth. This mechanical stress is transduced intracellularly via integrins, the Rho signaling pathway, and the Hippo signaling pathway, thereby promoting tumor invasion. Additionally, mechanical pressure fosters glycolysis in tumor cells, boosting energy production to support metastasis. Mechanical cues also regulate macrophage polarization, maintaining an inflammatory microenvironment conducive to tumor survival. In summary, mechanical signals within tumors play a crucial role in tumor growth and invasion. Understanding these signals and their involvement in tumor progression is essential for advancing our knowledge of tumor biology and enhancing therapeutic approaches.
Collapse
Affiliation(s)
- Yanli Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China.
| | - Qi Fu
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Wenyue Sun
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Qiujuan Yue
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Ping He
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China
| | - Dong Niu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Min Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi Province, China.
| |
Collapse
|
4
|
Li X, Chen Y, Li X, Yang X, Zhou L, Cheng Y, Hou H, Yang D, Gong Y, Xiao H, Wang J. Weight Management for Fertility-Preservation Therapy in Endometrial Cancer: Opportunities and Challenges. Curr Oncol Rep 2025; 27:195-210. [PMID: 39913071 DOI: 10.1007/s11912-025-01635-9] [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] [Accepted: 01/06/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE OF REVIEW Obesity is increasingly recognized as a significant factor impacting the outcomes of fertility-preserving therapies for endometrial cancer (EC). This review explores the effects of glycolipid metabolism on EC and its relationship with body weight. We will examine how excess body weight influences the effectiveness of fertility-preserving treatments and discuss potential mechanisms for effective weight management. Additionally, the review highlights the importance of comprehensive weight management as an adjunct strategy to enhance the efficacy of fertility-preserving interventions, providing insights into how to integrate metabolic health into clinical treatment protocols. RECENT FINDINGS Weight management can modify the tumor microenvironment by depriving the tumor of nutrients, whereas exercise can enhance immunity, potentially leading to tumor cell death. In addition, progesterone therapy may impede the proliferation of EC cells. Comprehensive weight management can serve as an essential adjuvant treatment for patients undergoing fertility-preserving therapies for EC. In this review, we highlight that comprehensive weight management can serve as a crucial adjuvant treatment for patients undergoing fertility-preserving therapies for endometrial cancer. Targeting glycolipid metabolism and addressing adiposity can improve hormonal balance, reduce inflammation, and enhance fertility outcomes. Further research is necessary to establish specific protocols and evaluate the effectiveness of these strategies in clinical practice.
Collapse
Affiliation(s)
- XiaoDan Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - YiQian Chen
- Beijing Health Vocational College, Beijing, 101101, China
| | - XiaoWei Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Yuan Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - HongYi Hou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Dandan Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Yuanyuan Gong
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Haihua Xiao
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
5
|
Bucurica S, Nancoff AS, Marin RI, Preda CM. Hepatocellular Carcinoma in Patients with Chronic Hepatitis C and Liver Cirrhosis Treated with DAA: A Focused Review. J Clin Med 2025; 14:1505. [PMID: 40095031 PMCID: PMC11900587 DOI: 10.3390/jcm14051505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: The issue of HCC recurrence in patients with liver cirrhosis and chronic HCV infection after DAA treatment as well as the issue of de novo HCC in individuals with chronic HCV hepatitis treated with DAA is of great importance. In this review, the two important aspects are discussed and, finally, an algorithm for approaching the patient with HCC and chronic HCV infection is proposed. Methods: A literature search of the two databases (PubMed and Scopus) was conducted using the terms 'chronic hepatitis C' and/or 'liver cirrhosis' and 'hepatocellular carcinoma', from database inception to December 2024. Results: Thirty-one studies have examined the risk of HCC recurrence. Most of these studies conclude that DAA treatment reduces the risk of HCC recurrence compared to patients who did not receive DAA. There are considerable differences across various world regions. These variations may arise from: differences in genotypes, baseline characteristics of the populations, variability in DAA treatment protocols, and differences in follow-up intervals. Eleven studies that investigated the issue of de novo HCC after DAA were reviewed, of which two included historical cohorts of untreated patients. Conclusions: The conclusion is that these patients present a low or equal risk of HCC incidence compared to untreated patients, and the risk factors for HCC are: lower platelet number, impaired liver function, nonresponse to DAA. Most patients with chronic hepatitis C and HCC should receive DAAs, except for those in BCLC stage D, but we must emphasize that timing of intervention is crucial and it is very important to evaluate possible drug interactions.
Collapse
Affiliation(s)
- Sandica Bucurica
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (S.B.); (A.-S.N.)
- Department of Gastroenterology, University Emergency Central Military Hospital “Dr. Carol Davila”, 024185 Bucharest, Romania
| | - Andreea-Simona Nancoff
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (S.B.); (A.-S.N.)
| | - Raluca Ioana Marin
- Department of Gastroenterology, Fundeni Clinic Institute, 022328 Bucharest, Romania;
| | - Carmen Monica Preda
- Department of Gastroenterology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (S.B.); (A.-S.N.)
- Department of Gastroenterology, Fundeni Clinic Institute, 022328 Bucharest, Romania;
| |
Collapse
|
6
|
Fan X, Chen H, Li Y, Feng Q, Tao F, Xu C, Chen X, Gao R, Wang Y, Guo X, Sun C, Lv C, Cheng Y. Actin-Targeted Magnetic Nanomotors Mechanically Modulate the Tumor Mechanical Microenvironment for Cancer Treatment. ACS NANO 2025; 19:6454-6467. [PMID: 39915111 DOI: 10.1021/acsnano.4c17229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
The abnormal mechanical microenvironment is a hallmark feature of solid tumors and plays a key role in immunotherapy resistance. The actin cytoskeleton can be finely tuned to control cell mechanics, which becomes a central target to regulate the tumor mechanical microenvironment (TMME). Here, we propose an actin-binding protein-modified magnetic nanomotor (ABP-MN) coupled with the rotating magnetic field (MF) to dynamically regulate the actin cytoskeleton for remodeling the TMME. ABP-MNs, with an ultrasmall diameter of 23 nm, intracellularly target the actin cytoskeleton and induce depolymerization via magneto-mechanical force under MF. Cancer-associated fibroblasts (CAFs) and tumor cells, which internalize ∼69.3% of ABP-MNs, are significantly tuned under MF with signs of a 7-fold decrease in tumor matrix stiffness, increased immune cell infiltration, and 95.8% tumor growth inhibition. This strategy unlocks a fresh field to reshape the TMME with the intracellular mechanical approach, thereby providing an effective mechano-based therapy in treating solid tumors.
Collapse
Affiliation(s)
- Xing Fan
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Haotian Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai 201210, China
| | - Yuan Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Qishuai Feng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Feng Tao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Chang Xu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xiaolei Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Rui Gao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yingying Wang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xinyu Guo
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Chenkai Sun
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Cheng Lv
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yu Cheng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Collaborative Innovation Center for Brain Science, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai 201210, China
| |
Collapse
|
7
|
Tandon R, Kumar S, Handa M, Srivastava N. Exosomes in glioma: mechanistic insights on biological, therapeutic, and diagnostic perspective. Ther Deliv 2025:1-12. [PMID: 39957239 DOI: 10.1080/20415990.2025.2466410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
Gliomas are prominent and frequent primary malignant brain tumors, with a generally poor prognosis. Current treatment involves radiation, surgery and chemotherapy. Exosomes are nanoscale extracellular vesicles released by cells that enable biological molecule movement and encourage intercellular communication in the tumor microenvironment. This contributes to glioma development, radiation resistance, and overcomes chemotherapy. Exosome functional and structural properties are essential for understanding cancer molecular mechanisms. They can also treat invasive tumors like glioblastomas and serve as diagnostic markers. Recent research depicted exosomes' prominent role in cancer cell maintenance, intercellular signaling, and microenvironment modification. Exosomes hold nucleic acids, proteins, lipids, mRNAs, lncRNAs, miRNAs, and immunological regulatory molecules depending on the origin of the cell. This paper reviews exosomes, their role in glioma etiology, and perspective diagnostic and therapeutic uses.
Collapse
Affiliation(s)
- Reetika Tandon
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| | - Samarth Kumar
- Formulation Research & Development-Non Orals, Sun Pharmaceuticals Industries Limited, Vadodara, India
| | - Mayank Handa
- Formulation Research & Development-Non Orals, Sun Pharmaceuticals Industries Limited, Vadodara, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| |
Collapse
|
8
|
Townson J, Progida C. The emerging roles of the endoplasmic reticulum in mechanosensing and mechanotransduction. J Cell Sci 2025; 138:JCS263503. [PMID: 39976266 DOI: 10.1242/jcs.263503] [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] [Indexed: 02/21/2025] Open
Abstract
Cells are continuously subjected to physical and chemical cues from the extracellular environment, and sense and respond to mechanical cues via mechanosensation and mechanotransduction. Although the role of the cytoskeleton in these processes is well known, the contribution of intracellular membranes has been long neglected. Recently, it has become evident that various organelles play active roles in both mechanosensing and mechanotransduction. In this Review, we focus on mechanosensitive roles of the endoplasmic reticulum (ER), the functions of which are crucial for maintaining cell homeostasis. We discuss the effects of mechanical stimuli on interactions between the ER, the cytoskeleton and other organelles; the role of the ER in intracellular Ca2+ signalling via mechanosensitive channels; and how the unfolded protein response and lipid homeostasis contribute to mechanosensing. The expansive structure of the ER positions it as a key intracellular communication hub, and we additionally explore how this may be leveraged to transduce mechanical signals around the cell. By synthesising current knowledge, we aim to shed light on the emerging roles of the ER in cellular mechanosensing and mechanotransduction.
Collapse
Affiliation(s)
- Jonathan Townson
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| |
Collapse
|
9
|
Li S, Chen Y, Feng S, Liu Z, Gan L, Wang Q. Autophagy-targeted Pt(IV) agents: a new horizon in antitumor drug development. Dalton Trans 2025; 54:1770-1778. [PMID: 39804164 DOI: 10.1039/d4dt03162h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Pt(IV) complexes as prodrugs of Pt(II) drugs exhibit numerous advantages such as enhanced stability, reduced toxicity, increased oral bioavailability, and efficacy in overcoming the drug resistance of Pt(II) compounds, which underscore their significant potential in the advancement of novel Pt anticancer agents. Furthermore, protective autophagy is pivotal in sustaining tumor cell homeostasis and modulating the tumor microenvironment (TME), thereby representing a critical target for the development of antitumor drugs. Specific inhibition or activation of autophagy during chemotherapy would break the internal homeostasis in the TME and increase antitumor activities. Consequently, developing novel Pt(IV) antitumor agents with autophagy-targeting capabilities by incorporating autophagy-regulating moieties into the Pt(IV) framework has emerged as a hot topic in the discovery of novel Pt drugs. Herein, the research progress in novel Pt(IV) antitumor drugs with autophagy-targeted properties is systematically reviewed based on the literature. The future challenges and perspectives of this fascinating class of conjugates are also discussed, aiming to provide new insights and approaches for the future design and investigation of novel Pt antitumor drugs.
Collapse
Affiliation(s)
- Suying Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, P.R. China
| | - Shuaiqi Feng
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Linling Gan
- Chongqing Engineering Research Center of Pharmaceutical Sciences, School of Pharmacy, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, P. R. China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| |
Collapse
|
10
|
Dar MS, Rosaiah P, Bhagyalakshmi J, Ahirwar S, Khan A, Tamizhselvi R, Reddy VRM, Palaniappan A, Sahu NK. Graphene quantum dots as nanotherapeutic agents for triple-negative breast cancer: Insights from 3D tumor models. Coord Chem Rev 2025; 523:216247. [DOI: 10.1016/j.ccr.2024.216247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Cai G, Rodgers NC, Liu AP. Unjamming Transition as a Paradigm for Biomechanical Control of Cancer Metastasis. Cytoskeleton (Hoboken) 2024. [PMID: 39633605 DOI: 10.1002/cm.21963] [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: 08/14/2024] [Revised: 10/27/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Tumor metastasis is a complex phenomenon that poses significant challenges to current cancer therapeutics. While the biochemical signaling involved in promoting motile phenotypes is well understood, the role of biomechanical interactions has recently begun to be incorporated into models of tumor cell migration. Specifically, we propose the unjamming transition, adapted from physical paradigms describing the behavior of granular materials, to better discern the transition toward an invasive phenotype. In this review, we introduce the jamming transition broadly and narrow our discussion to the different modes of 3D tumor cell migration that arise. Then we discuss the mechanical interactions between tumor cells and their neighbors, along with the interactions between tumor cells and the surrounding extracellular matrix. We center our discussion on the interactions that induce a motile state or unjamming transition in these contexts. By considering the interplay between biochemical and biomechanical signaling in tumor cell migration, we can advance our understanding of biomechanical control in cancer metastasis.
Collapse
Affiliation(s)
- Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole C Rodgers
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Allen P Liu
- Applied Physics Program, University of Michigan, Ann Arbor, Michigan, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Gargalionis AN, Papavassiliou KA, Basdra EK, Papavassiliou AG. Advances in non-small cell lung cancer mechanomedicine: deciphering the signaling networks that govern tumor-TME interactions. J Exp Clin Cancer Res 2024; 43:316. [PMID: 39616383 PMCID: PMC11608457 DOI: 10.1186/s13046-024-03242-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 11/28/2024] [Indexed: 03/21/2025] Open
Abstract
Cells from the tumor microenvironment (TME) interact with tumor cells in non-small cell lung cancer (NSCLC) to form a reciprocal crosstalk which influences tumor growth, proliferation, metastasis and multidrug response. This crosstalk is modulated by TME mechanical inputs, which elicit the processes of mechanosensing and mechanotransduction. Recent advances in unveiling these signaling networks establish the interdisciplinary field of mechanomedicine to exploit emerging diagnostic, predictive and therapeutic tools for more effective NSCLC treatments.
Collapse
Affiliation(s)
- Antonios N Gargalionis
- Laboratory of Clinical Biochemistry, Medical School, 'Attikon' University General Hospital, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Kostas A Papavassiliou
- First University Department of Respiratory Medicine, Medical School, 'Sotiria' Chest Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Efthimia K Basdra
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| |
Collapse
|
13
|
Nie Y, Lu X, Zhu Y, Shi Y, Ren K, Li Z, Chen P, Han D, Li X. Circular Adhesion Substrates Inhibiting Cell Polarization and Proliferation via Graded Texture of Geometric Micropatterns. SMALL METHODS 2024:e2401471. [PMID: 39564718 DOI: 10.1002/smtd.202401471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/06/2024] [Indexed: 11/21/2024]
Abstract
Most melanomas that occur on the skin surface originate from a newly formed nevus and grow outward in a circular pattern and metastasize from the nevus center. Herein, a circular microfabricated substrate is constructed to explore the growth behavior of melanoma cells. Modeling software is used to calculate appropriate parameters, including shape and size, and then the substrates are processed with microfabrication technologies. The results show that the melanoma cells on the circular adhesion substrate are oval and are significant changes in cell spread length, nuclei, area, aspect ratio, Young's modulus, and orientation angles, indicating inhibition of cell polarization. Moreover, three different layers from circular adhesion substrates are selected to construct new substrates, which indicates that the polarization degree of cells is closely related to the number of micropillar arrays on the circular geometric substrate. In addition, flow cytometry demonstrates that the circular substrate reduced the transition from resting/gap 1 phase (G0/G1) to synthesis phase (S phase), thereby decreasing DNA synthesis and proliferation, reminding a potential method for treatment strategy. More importantly, the circular adhesion substrate influences the integrin signaling pathway, which has a potential application and research prospect in the treatment of melanoma.
Collapse
Affiliation(s)
- Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Xi Lu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, P. R. China
| | - Yuting Zhu
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
| | - Yahong Shi
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, P. R. China
| | - Keli Ren
- The Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhongxian Li
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Peipei Chen
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Dong Han
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiang Li
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| |
Collapse
|
14
|
Wang H, Li Q, Tang Q, Shi G, Wu G, Mao X, Wu C, Zhang L, Liu J, Li J, Li B. Role and therapeutic potential of E3s in the tumor microenvironment of hepatocellular carcinoma. Front Immunol 2024; 15:1483721. [PMID: 39544935 PMCID: PMC11560419 DOI: 10.3389/fimmu.2024.1483721] [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: 08/20/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a high-incidence, poor-prognosis malignancy worldwide, requiring new strategies for treatment. Ubiquitination, especially ubiquitination through E3 ubiquitin ligases, plays an indispensable role in the development and progression of HCC. E3 ubiquitin ligases are crucial enzymes in ubiquitination, controlling the degradation of specific substrate proteins and influencing various cellular functions, such as tumor cell proliferation, apoptosis, migration, and immune evasion. In this review, we systematically summarize the mechanisms of E3 ubiquitin ligases in HCC, with a focus on the significance of RING, HECT, and RBR types in HCC progression. The review also looks at the potential for targeting E3 ligases to modulate the tumor microenvironment (TME) and increase immunotherapy efficacy. Future studies will optimize HCC treatment by formulating specific inhibitors or approaches that will be based on gene therapy targeting E3 ligases in order to overcome resistance issues with present treatments and create optimism in the journey of treatment for HCC patients.
Collapse
Affiliation(s)
- Hailin Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiang Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qinqin Tang
- Department of Dermatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Gang Shi
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Guo Wu
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xingbo Mao
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Changkang Wu
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lixin Zhang
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jingdong Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Wu M, Liao Y, Tang L. Non-small cell lung cancer organoids: Advances and challenges in current applications. Chin J Cancer Res 2024; 36:455-473. [PMID: 39539817 PMCID: PMC11555200 DOI: 10.21147/j.issn.1000-9604.2024.05.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Lung cancer is emerging as a common malignancy worldwide, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all cases. Two-dimensional (2D) in vitro cell line cultures and animal models are currently used to study NSCLC. However, 2D cell cultures fail to replicate the medication response and neoplastic heterogeneity of parental tumors. Animal models are expensive and require lengthy modeling cycles. The generation of in vitro three-dimensional (3D) tissue cultures called organoids, which exhibit multicellular, anatomical, and functional properties of real organs, is now achievable owing to advancements in stem cell culturing. The genetic, proteomic, morphological, and pharmacological characteristics of tumors are largely preserved in tumor organoids grown in vitro. The design and physiology of human organs can be precisely reconstructed in tumor organoids, opening new possibilities for complementing the use of animal models and studying human diseases. This review summarizes the development of NSCLC organoids and their applications in basic research, drug testing, immunotherapy, and individualized treatments.
Collapse
Affiliation(s)
- Maoqin Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yi Liao
- Department of Technical Support, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning 530021, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
16
|
Chen Y, Zhang M, Liu Z, Zhang N, Wang Q. Ursodeoxycholic Acid Platinum(IV) Conjugates as Antiproliferative and Antimetastatic Agents: Remodel the Tumor Microenvironment through Suppressing JAK2/STAT3 Signaling. J Med Chem 2024; 67:17551-17567. [PMID: 39292635 DOI: 10.1021/acs.jmedchem.4c01549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Tumor microenvironment (TME) is a pivotal factor driving the tumor metastasis and leading to the failure of tumor therapy. Here, a series of ursodeoxycholic acid platinum(IV) conjugates with potency in remodeling the TME through suppressing JAK2/STAT3 signaling was developed. A candidate was screened out, which displayed potent antiproliferative and antimetastatic performance both in vitro and in vivo. It displayed superior pharmacokinetic properties compared to cisplatin. Serious DNA injury was induced, and then mitochondria-mediated apoptosis was initiated through the Bcl-2/Bax/Caspase3 pathway. The JAK2/STAT3 and TGF-β1 signaling pathways were remarkably inhibited, and pro-death autophagy was subsequently promoted. The inflammatory and hypoxic TME was suppressed by downregulating COX-2, MMP9, and HIF-1α, which resulted in inhibited angiogenesis in tumors by inhibiting the HIF-1α/VEGFA axis. Additionally, the immunosuppressive TME was reversed by blocking the immune checkpoint PD-L1, further improving the density of CD3+ and CD8+ tumor-infiltrating lymphocytes, and promoting macrophage polarization from M2- to M1-type.
Collapse
Affiliation(s)
- Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Ming Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China
| |
Collapse
|
17
|
Bareham B, Dibble M, Parsons M. Defining and modeling dynamic spatial heterogeneity within tumor microenvironments. Curr Opin Cell Biol 2024; 90:102422. [PMID: 39216233 PMCID: PMC11846781 DOI: 10.1016/j.ceb.2024.102422] [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: 05/08/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Many solid tumors exhibit significant genetic, cellular, and biophysical heterogeneity which dynamically evolves during disease progression and after treatment. This constant flux in cell composition, phenotype, spatial relationships, and tissue properties poses significant challenges in accurately diagnosing and treating patients. Much of the complexity lies in unraveling the molecular changes in different tumor compartments, how they influence one another in space and time and where vulnerabilities exist that might be appropriate to target therapeutically. Recent advances in spatial profiling tools and technologies are enabling new insight into the underlying biology of complex tumors, creating a greater understanding of the intricate relationship between cell types, states, and the microenvironment. Here we reflect on some recent discoveries in this area, where the key knowledge and technology gaps lie, and the advancements in spatial measurements and in vitro models for the study of spatial intratumoral heterogeneity.
Collapse
Affiliation(s)
- Bethany Bareham
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Matthew Dibble
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
18
|
Niculet E, Radaschin DS, Arbune M, Bobeica C, Craescu M, Onisor C, Gurau G, Busila C, Vasile CI, Tatu AL. Basal Cell Carcinoma Cleft: The Missing Piece of the Puzzle. Cureus 2024; 16:e71244. [PMID: 39525230 PMCID: PMC11550456 DOI: 10.7759/cureus.71244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This study aims to explore the tumor-stroma separation or the cleft characterizing basal cell carcinoma (BCC). METHODOLOGY In this retrospective cohort investigation, we enrolled 244 patients who received a confirmed diagnosis of BCC through histopathological examination in the period of 2019-2020 at the Pathology Laboratory of the "Sfântul Apostol Andrei" Emergency Clinical Hospital located in Galați, Romania. The identification of patients was accomplished by utilizing electronic health records, and relevant clinical, demographic, and histopathological data were retrieved from the physical database of the Pathology Laboratory. Key tumor characteristics were gathered, and an in-depth analysis of case slides was performed. RESULTS The average tumor-stroma cleft's width measurement was 48.136 µm, while its respective tumor island's width was on average 952.587 µm. The cleft's width and its respective tumor island's width are dependent on the BCC subtype, just like the ratio between the tumor island's measurement and its cleft's width are, being larger in basosquamous BCC, micronodular BCC, infiltrative BCC, and morpheaform BCC. CONCLUSION The BCC tumor islands were found to have a minimal approximately equal measurement to the tumor-stroma separation cleft, but they were always larger than the latter. Large clefts and their respective tumor islands were found in specific tumor subtypes such as basosquamous BCC, micronodular BCC, infiltrative BCC, and morpheaform BCC, but in nodular BCC also.
Collapse
Affiliation(s)
- Elena Niculet
- Department of Pathology, "Sfantul Apostol Andrei" Emergency Clinical Hospital of Galati, Romania, Galati, ROU
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR), "Dunărea de Jos" University, Galati, ROU
| | - Diana S Radaschin
- Department of Dermatology, Saint Parascheva Infectious Disease Clinical Hospital, Galati, ROU
- Biomedical Doctoral School, "Dunărea de Jos" University, Galati, ROU
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR), "Dunărea de Jos" University, Galati, ROU
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Manuela Arbune
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Carmen Bobeica
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Mihaela Craescu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Cristian Onisor
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Gabriela Gurau
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Camelia Busila
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galați, ROU
| | - Claudiu I Vasile
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Alin L Tatu
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR), "Dunărea de Jos" University, Galati, ROU
- Department of Dermatology, "Sf. Cuvioasa Parascheva" Clinical Hospital of Infectious Diseases, Galati, ROU
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| |
Collapse
|
19
|
Hong J, Yu J, Buratto D, Chen W, Zhou R, Ling S, Xu X. Unveiling the Role of Mechanical Microenvironment in Hepatocellular Carcinoma: Molecular Mechanisms and Implications for Therapeutic Strategies. Int J Biol Sci 2024; 20:5239-5253. [PMID: 39430235 PMCID: PMC11489173 DOI: 10.7150/ijbs.102706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 09/15/2024] [Indexed: 10/22/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer in the world and the third leading cause of cancer deaths globally. More than 80% of HCC patients have a background of fibrosis or cirrhosis, which leads to changes in physical factors in tumor microenvironment (TME), such as increased stiffness, solid stress, fluid stresses and structural alterations in the extracellular matrix (ECM). In the past, the focus of cancer research has predominantly been on genetic and biochemical factors in the TME, and the critical role of physical factors has often been overlooked. Recent discoveries suggest these unique physical signals are converted into biochemical signals through a mechanotransduction process that influences the biological behavior of tumor cells and stromal cells. This process facilitates the occurrence and progression of tumors. This review delves into the alterations in the mechanical microenvironment during the progression of liver fibrosis to HCC, the signaling pathways activated by physical signals, and the effects on both tumor and mesenchymal stromal cells. Furthermore, this paper summarizes and discusses the therapeutic options for targeting the mechanical aspects of the TME, offering valuable insights for future research into novel therapeutic avenues against HCC and other solid tumors.
Collapse
Affiliation(s)
- Jiachen Hong
- Hangzhou Normal University, Hangzhou, 311121, China
| | - Jiongjie Yu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, China
| | - Damiano Buratto
- Institute of Quantitative Biology, and College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
| | - Wei Chen
- Department of Cell Biology, Zhejiang University School of Medicine, and Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, and College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Sunbin Ling
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou 314408, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou 314408, China
- The Second Clinical College of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
20
|
Li Z, Li J, Bai X, Huang X, Wang Q. Tumor microenvironment as a complex milieu driving cancer progression: a mini review. Clin Transl Oncol 2024:10.1007/s12094-024-03697-w. [PMID: 39342061 DOI: 10.1007/s12094-024-03697-w] [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: 03/29/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024]
Abstract
It has been spotlighted that the Tumor Microenvironment (TME) is crucial for comprehending cancer progression and therapeutic resistance. Therefore, this comprehensive review elucidates the intricate architecture of the TME, which encompasses tumor cells, immune components, support cells, and a myriad of bioactive molecules. These constituents collectively foster dynamic interactions that underpin tumor growth, metastasis, and nuanced responses to anticancer therapies. Notably, the TME's role extends beyond mere physical support, serving as a critical mediator in cancer-cell evolution, immune modulation, and treatment outcomes. Innovations targeting the TME, including strategies focused on the vasculature, immune checkpoints, and T-cell therapies, have forged new pathways for clinical intervention. However, the heterogeneity and complexity of the TME present significant challenges, necessitating deeper exploration of its components and their interplay to enhance therapeutic efficacy. This review underscores the imperative for integrated research strategies that amalgamate insights from tumor biology, immunology, and systems biology. Such an approach aims to refine cancer treatments and improve patient prognoses by exploiting the TME's complexity.
Collapse
Affiliation(s)
- Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Center for Stomatology, Shanghai, China.
- National Clinical Research Center for Oral Diseases, Shanghai, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, China.
- Shanghai Research Institute of Stomatology, Shanghai, China.
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, China.
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Jing Li
- Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
| | - Xiaolei Bai
- Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary.
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
21
|
Tan M, Song B, Zhao X, Du J. The role and mechanism of compressive stress in tumor. Front Oncol 2024; 14:1459313. [PMID: 39351360 PMCID: PMC11439826 DOI: 10.3389/fonc.2024.1459313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Recent research has revealed the important role of mechanical forces in the initiation and progression of tumors. The interplay between mechanical and biochemical cues affects the function and behavior of tumor cells during the development of solid tumors, especially their metastatic potential. The compression force generated by excessive cell proliferation and the tumor microenvironment widely regulates the progression of solid tumor disease. Tumor cells can sense alterations in compressive stress through diverse mechanosensitive components and adapt their mechanical characteristics accordingly to adapt to environmental changes. Here, we summarize the current role of compressive stress in regulating tumor behavior and its biophysical mechanism from the mechanobiological direction.
Collapse
Affiliation(s)
- Min Tan
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Bingqi Song
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xinbin Zhao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
| | - Jing Du
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
22
|
Xiao C, Xie N, Shu Q, Liang X, Wang Z, Wu J, Shi N, Huang X, Wei ZC, Gao X, Liu H, Wu K, Xu J, Wang JH, Liu N, Xu F. Synergistic Effects of Matrix Biophysical Properties on Gastric Cancer Cell Behavior via Integrin-Mediated Cell-ECM Interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309907. [PMID: 38712486 DOI: 10.1002/smll.202309907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
The biophysical properties of the extracellular matrix (ECM) play a pivotal role in modulating cancer progression via cell-ECM interactions. However, the biophysical properties specific to gastric cancer (GC) remain largely unexplored. Pertinently, GC ECM shows significantly heterogeneous metamorphoses, such as matrix stiffening and intricate restructuring. By combining collagen I and alginate, this study designs an in vitro biomimetic hydrogel platform to independently modulate matrix stiffness and structure across a physiological stiffness spectrum while preserving consistent collagen concentration and fiber topography. With this platform, this study assesses the impacts of matrix biophysical properties on cell proliferation, migration, invasion, and other pivotal dynamics of AGS. The findings spotlight a compelling interplay between matrix stiffness and structure, influencing both cellular responses and ECM remodeling. Furthermore, this investigation into the integrin/actin-collagen interplay reinforces the central role of integrins in mediating cell-ECM interactions, reciprocally sculpting cell conduct, and ECM adaptation. Collectively, this study reveals a previously unidentified role of ECM biophysical properties in GC malignant potential and provides insight into the bidirectional mechanical cell-ECM interactions, which may facilitate the development of novel therapeutic horizons.
Collapse
Affiliation(s)
- Cailan Xiao
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Ning Xie
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Qiuai Shu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Xiru Liang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Ziwei Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Jian Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Nianyuan Shi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Key Laboratory of Magnetic Medicine, Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xindi Huang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Zhong-Cao Wei
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Xiaoliang Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Hao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Kaichun Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Jingyuan Xu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, P. R. China
| | - Jin-Hai Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Na Liu
- Department of Gastroenterology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| |
Collapse
|
23
|
Yang P, Jiang Y, Chen R, Yang J, Liu M, Huang X, Xu G, Hao R. Prognostic and immune infiltration implications of SIGLEC9 in SKCM. Diagn Pathol 2024; 19:112. [PMID: 39153970 PMCID: PMC11330613 DOI: 10.1186/s13000-024-01536-8] [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: 06/25/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024] Open
Abstract
The occurrence and progression of skin cutaneous melanoma (SKCM) is strongly associated with immune cells infiltrating the tumor microenvironment (TME). This study examined the expression, prognosis, and immune relevance of SIGLEC9 in SKCM using multiple online databases. Analysis of the GEPIA2 and Ualcan databases revealed that SIGLEC9 is highly expressed in SKCM, and patients with high SIGLEC9 expression had improved overall survival (OS). Furthermore, the mutation rate of SIGLEC9 in SKCM patients was found to be 5.41%, the highest observed. The expression of SIGLEC9 was positively correlated with macrophages, neutrophils and B cells, CD8 + T cells, CD4 + T cells, and dendritic cells, according to TIMER. Based on TCGA-SKCM data, we verified that high SIGLEC9 expression is closely associated with a good prognosis for SKCM patients, including overall survival, progression-free interval, and disease-specific survival. This positive prognosis could be due to the infiltration of immune cells into the TME. Additionally, our analysis of single-cell transcriptome data revealed that SIGLEC9 not only played a role in the normal skin immune microenvironment, but is also highly expressed in immune cell subpopulations of SKCM patients, regulating the immune response to tumors. Our findings suggest that the close association between SIGLEC9 and SKCM prognosis is primarily mediated by its effect on the tumor immune microenvironment.
Collapse
Affiliation(s)
- Peipei Yang
- Department of Dermatology, Jingmen People'S Hospital &, Jingchu University of Technology Affiliated Central Hospital, Jingmen, 448000, China
| | - Yunhui Jiang
- Department of Pathology, Jingmen People'S Hospital &, Jingchu University of Technology Affiliated Central Hospital, Jingmen, 448000, China
| | - Rong Chen
- Department of Clinical Laboratory, Jingmen People'S Hospital &, Jingchu University of Technology Affiliated Central Hospital, Jingmen, 448000, China
| | - Junhan Yang
- Department of Dermatology, Jingmen People'S Hospital &, Jingchu University of Technology Affiliated Central Hospital, Jingmen, 448000, China
| | - Mengting Liu
- Department of Dermatology, Jingmen People'S Hospital &, Jingchu University of Technology Affiliated Central Hospital, Jingmen, 448000, China
| | - Xieping Huang
- Department of Dermatology, Jingmen People'S Hospital &, Jingchu University of Technology Affiliated Central Hospital, Jingmen, 448000, China
| | - Ganglin Xu
- Department of Dermatology, Jingmen People'S Hospital &, Jingchu University of Technology Affiliated Central Hospital, Jingmen, 448000, China.
| | - Rui Hao
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, China.
| |
Collapse
|
24
|
Aydin H, Ozcelikkale A, Acar A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater Sci Eng 2024; 10:4682-4700. [PMID: 38967485 PMCID: PMC11322920 DOI: 10.1021/acsbiomaterials.4c00445] [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: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.
Collapse
Affiliation(s)
- Hakan
Berk Aydin
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| | - Altug Ozcelikkale
- Department
of Mechanical Engineering, Middle East Technical
University, 06800, Ankara, Turkey
- Graduate
Program of Biomedical Engineering, Middle
East Technical University, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| |
Collapse
|
25
|
Brooks A, Zhang Y, Chen J, Zhao CX. Cancer Metastasis-on-a-Chip for Modeling Metastatic Cascade and Drug Screening. Adv Healthc Mater 2024; 13:e2302436. [PMID: 38224141 DOI: 10.1002/adhm.202302436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Microfluidic chips are valuable tools for studying intricate cellular and cell-microenvironment interactions. Traditional in vitro cancer models lack accuracy in mimicking the complexities of in vivo tumor microenvironment. However, cancer-metastasis-on-a-chip (CMoC) models combine the advantages of 3D cultures and microfluidic technology, serving as powerful platforms for exploring cancer mechanisms and facilitating drug screening. These chips are able to compartmentalize the metastatic cascade, deepening the understanding of its underlying mechanisms. This article provides an overview of current CMoC models, focusing on distinctive models that simulate invasion, intravasation, circulation, extravasation, and colonization, and their applications in drug screening. Furthermore, challenges faced by CMoC and microfluidic technologies are discussed, while exploring promising future directions in cancer research. The ongoing development and integration of these models into cancer studies are expected to drive transformative advancements in the field.
Collapse
Affiliation(s)
- Anastasia Brooks
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Yali Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Jiezhong Chen
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
26
|
Zubiarrain-Laserna A, Martínez-Moreno D, López de Andrés J, de Lara-Peña L, Guaresti O, Zaldua AM, Jiménez G, Marchal JA. Beyond stiffness: deciphering the role of viscoelasticity in cancer evolution and treatment response. Biofabrication 2024; 16:042002. [PMID: 38862006 DOI: 10.1088/1758-5090/ad5705] [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: 11/23/2023] [Accepted: 06/11/2024] [Indexed: 06/13/2024]
Abstract
There is increasing evidence that cancer progression is linked to tissue viscoelasticity, which challenges the commonly accepted notion that stiffness is the main mechanical hallmark of cancer. However, this new insight has not reached widespread clinical use, as most clinical trials focus on the application of tissue elasticity and stiffness in diagnostic, therapeutic, and surgical planning. Therefore, there is a need to advance the fundamental understanding of the effect of viscoelasticity on cancer progression, to develop novel mechanical biomarkers of clinical significance. Tissue viscoelasticity is largely determined by the extracellular matrix (ECM), which can be simulatedin vitrousing hydrogel-based platforms. Since the mechanical properties of hydrogels can be easily adjusted by changing parameters such as molecular weight and crosslinking type, they provide a platform to systematically study the relationship between ECM viscoelasticity and cancer progression. This review begins with an overview of cancer viscoelasticity, describing how tumor cells interact with biophysical signals in their environment, how they contribute to tumor viscoelasticity, and how this translates into cancer progression. Next, an overview of clinical trials focused on measuring biomechanical properties of tumors is presented, highlighting the biomechanical properties utilized for cancer diagnosis and monitoring. Finally, this review examines the use of biofabricated tumor models for studying the impact of ECM viscoelasticity on cancer behavior and progression and it explores potential avenues for future research on the production of more sophisticated and biomimetic tumor models, as well as their mechanical evaluation.
Collapse
Affiliation(s)
- Ana Zubiarrain-Laserna
- Leartiker S. Coop., Xemein Etorbidea 12A, 48270 Markina-Xemein, Spain
- BioFab i3D- Biofabrication and 3D (bio)printing Laboratory, University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM) University of Granada, Granada, Spain
| | - Daniel Martínez-Moreno
- BioFab i3D- Biofabrication and 3D (bio)printing Laboratory, University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM) University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Excellence Research Unit 'Modeling Nature' (MNat), University of Granada, Granada, Spain
| | - Julia López de Andrés
- BioFab i3D- Biofabrication and 3D (bio)printing Laboratory, University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM) University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Excellence Research Unit 'Modeling Nature' (MNat), University of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Laura de Lara-Peña
- BioFab i3D- Biofabrication and 3D (bio)printing Laboratory, University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM) University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Excellence Research Unit 'Modeling Nature' (MNat), University of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Olatz Guaresti
- Leartiker S. Coop., Xemein Etorbidea 12A, 48270 Markina-Xemein, Spain
| | - Ane Miren Zaldua
- Leartiker S. Coop., Xemein Etorbidea 12A, 48270 Markina-Xemein, Spain
| | - Gema Jiménez
- BioFab i3D- Biofabrication and 3D (bio)printing Laboratory, University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM) University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Excellence Research Unit 'Modeling Nature' (MNat), University of Granada, Granada, Spain
- Department of Health Science, Faculty of Experimental Science, University of Jaen, 23071 Jaen, Spain
| | - Juan Antonio Marchal
- BioFab i3D- Biofabrication and 3D (bio)printing Laboratory, University of Granada, 18100 Granada, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM) University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Excellence Research Unit 'Modeling Nature' (MNat), University of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
27
|
Yu A, Zhang W, Zhang Q, Yang K, Liu X, Liu H, Xie J, Feng Y, Li J, Jia C. A TICT-AIE activated dual-channel fluorescence-on probe to reveal the dynamics mechanosensing of lipid droplets during ferroptosis. Talanta 2024; 274:126028. [PMID: 38599126 DOI: 10.1016/j.talanta.2024.126028] [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: 09/07/2023] [Revised: 01/18/2024] [Accepted: 03/30/2024] [Indexed: 04/12/2024]
Abstract
Mechanical forces play a crucial role in cellular processes, including ferroptosis, a form of regulated cell death associated with various diseases. However, the mechanical aspects of organelle lipid droplets (LDs) during ferroptosis are poorly understood. In this study, we designed and synthesized a fluorescent probe, TPE-V1, to enable real-time monitoring of LDs' viscosity using a dual-channel fluorescence-on model (red channel at 617 nm and NIR channel at 710 nm). The fluorescent imaging of using TPE-V1 was achieved due to the integrated mechanisms of the twisted intramolecular charge transfer (TICT) and aggregation-induced emission (AIE). Through dual-emission channel fluorescence imaging, we observed the enhanced mechanical energy of LDs triggering cellular mechanosensing, including ferroptosis and cell deformation. Theoretical calculations confirmed the probe's behavior, showing that high-viscosity media prevented the rotation processes and restored fluorescence quenching in low viscosity. These findings suggest that our TICT-TPE design strategy provides a practical approach to study LDs' mechanical properties during ferroptosis. This development enhances our understanding of the interplay between mechanical forces and LDs, contributing to the knowledge of ferroptotic cell death and potential therapeutic interventions targeting dysregulated cell death processes.
Collapse
Affiliation(s)
- Ao Yu
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China
| | - Wei Zhang
- Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199, China
| | - Qiangsheng Zhang
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China
| | - Kunlong Yang
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China
| | - Xiongbo Liu
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China
| | - Hongtao Liu
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, 570228, China
| | - Jialin Xie
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, 570228, China
| | - Yan Feng
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, 570228, China.
| | - Jianwei Li
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, Turku, 20520, Finland.
| | - Chunman Jia
- Hainan Provincial Key Laboratory of Fine Chem, School of Chemistry and Chemical Engineering, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, 570228, China; Analytical & Testing Center, Hainan University, Haikou, 570228, China.
| |
Collapse
|
28
|
Tseng YT, Tsai CC, Chen PC, Lin BY, Hsu SCN, Huang SP, Huang B. Mechanical shear flow regulates the malignancy of colorectal cancer cells. Kaohsiung J Med Sci 2024; 40:650-659. [PMID: 38757734 DOI: 10.1002/kjm2.12844] [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: 02/20/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Colorectal cancer (CRC) is notable for its high mortality and high metastatic characteristics. The shear force generated by bloodstream provides mechanical signals regulating multiple responses of cells, including metastatic cancer cells, dispersing in blood vessels. We, therefore, studied the effect of shear flow on circulating CRC cells in the present study. The CRC cell line SW620 was subjected to shear flow of 12.5 dynes/cm2 for 1 and 2 h separately. Resulting elevated caspase-9 and -3 indicated that shear flow initiated the apoptosis of SW620. Enlarged cell size associated with a higher level of cyclin D1 was coincident with the flow cytometric results indicating that the cell cycle was arrested at the G1 phase. An elevated phosphor-eNOSS1177 increased the production of nitric oxide and led to reactive oxygen species-mediated oxidative stress. Shear flow also regulated epithelial-mesenchymal transition (EMT) by increasing E-cadherin and ZO-1 while decreasing Snail and Twist1. The migration and invasion of sheared SW620 were also substantially decreased. Further investigations showed that mitochondrial membrane potential was significantly decreased, whereas mitochondrial mass and ATP production were not changed. In addition to the shear flow of 12.5 dynes/cm2, the expressions of EMT were compared at lower (6.25 dynes/cm2) and at higher (25 dynes/cm2) shear flow. The results showed that lower shear flow increased mesenchymal characteristics and higher shear flow increased epithelial characteristics. Shear flow reduces the malignancy of CRC in their metastatic dispersal that opens up new ways to improve cancer therapies by applying a mechanical shear flow device.
Collapse
Affiliation(s)
- Yu-Ting Tseng
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Chung Tsai
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Pediatrics, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Ping-Chen Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Bo-Yan Lin
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Ping Huang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Bin Huang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
29
|
Yang D, Dang S, Wang Z, Xie M, Li X, Ding X. Vessel co-option: a unique vascular-immune niche in liver cancer. Front Oncol 2024; 14:1386772. [PMID: 38737903 PMCID: PMC11082301 DOI: 10.3389/fonc.2024.1386772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Tumor vasculature is pivotal in regulating tumor perfusion, immune cell infiltration, metastasis, and invasion. The vascular status of the tumor is intricately linked to its immune landscape and response to immunotherapy. Vessel co-option means that tumor tissue adeptly exploits pre-existing blood vessels in the para-carcinoma region to foster its growth rather than inducing angiogenesis. It emerges as a significant mechanism contributing to anti-angiogenic therapy resistance. Different from angiogenic tumors, vessel co-option presents a distinctive vascular-immune niche characterized by varying states and distribution of immune cells, including T-cells, tumor-associated macrophages, neutrophils, and hepatic stellate cells. This unique composition contributes to an immunosuppressive tumor microenvironment that is crucial in modulating the response to cancer immunotherapy. In this review, we systematically reviewed the evidence and molecular mechanisms of vessel co-option in liver cancer, while also exploring its implications for anti-angiogenic drug resistance and the immune microenvironment, to provide new ideas and clues for screening patients with liver cancer who are effective in immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiangming Ding
- Department of Gastroenterology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| |
Collapse
|
30
|
Lightsey S, Sharma B. Natural Killer Cell Mechanosensing in Solid Tumors. Bioengineering (Basel) 2024; 11:328. [PMID: 38671750 PMCID: PMC11048000 DOI: 10.3390/bioengineering11040328] [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: 03/01/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Natural killer (NK) cells, which are an exciting alternative cell source for cancer immunotherapies, must sense and respond to their physical environment to traffic to and eliminate cancer cells. Herein, we review the mechanisms by which NK cells receive mechanical signals and explore recent key findings regarding the impact of the physical characteristics of solid tumors on NK cell functions. Data suggest that different mechanical stresses present in solid tumors facilitate NK cell functions, especially infiltration and degranulation. Moreover, we review recent engineering advances that can be used to systemically study the role of mechanical forces on NK cell activity. Understanding the mechanisms by which NK cells interpret their environment presents potential targets to enhance NK cell immunotherapies for the treatment of solid tumors.
Collapse
Affiliation(s)
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 23610, USA;
| |
Collapse
|
31
|
Wendong Y, Jiali J, Qiaomei F, Yayun W, Xianze X, Zheng S, Wei H. Biomechanical forces and force-triggered drug delivery in tumor neovascularization. Biomed Pharmacother 2024; 171:116117. [PMID: 38171243 DOI: 10.1016/j.biopha.2023.116117] [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: 11/02/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
Tumor angiogenesis is one of the typical hallmarks of tumor occurrence and development, and tumor neovascularization also exhibits distinct characteristics from normal blood vessels. As the number of cells and matrix inside the tumor increases, the biomechanical force is enhanced, specifically manifested as solid stress, fluid stress, stiffness, and topology. This mechanical microenvironment also provides shelter for tumors and intensifies angiogenesis, providing oxygen and nutritional support for tumor progression. During tumor development, the biomechanical microenvironment also emerges, which in turn feeds back to regulate the tumor progression, including tumor angiogenesis, and biochemical and biomechanical signals can regulate tumor angiogenesis. Blood vessels possess inherent sensitivity to mechanical stimuli, but compared to the extensive research on biochemical signal regulation, the study of the regulation of tumor neovascularization by biomechanical signals remains relatively scarce. Biomechanical forces can affect the phenotypic characteristics and mechanical signaling pathways of tumor blood vessels, directly regulating angiogenesis. Meanwhile, they can indirectly regulate tumor angiogenesis by causing an imbalance in angiogenesis signals and affecting stromal cell function. Understanding the regulatory mechanism of biomechanical forces in tumor angiogenesis is beneficial for better identifying and even taming the mechanical forces involved in angiogenesis, providing new therapeutic targets for tumor vascular normalization. Therefore, we summarized the composition of biomechanical forces and their direct or indirect regulation of tumor neovascularization. In addition, this review discussed the use of biomechanical forces in combination with anti-angiogenic therapies for the treatment of tumors, and biomechanical forces triggered delivery systems.
Collapse
Affiliation(s)
- Yao Wendong
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Jiang Jiali
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Fan Qiaomei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Weng Yayun
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Xie Xianze
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Shi Zheng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| | - Huang Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| |
Collapse
|
32
|
龙 世, 吴 翠, 曾 柱. [The Three-dimensional Environment of Type Ⅰ Collagen Gels With Varying Stiffness Modulates the Immunological Functions of NK Cells]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:81-86. [PMID: 38322517 PMCID: PMC10839474 DOI: 10.12182/20240160401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Indexed: 02/08/2024]
Abstract
Objective To construct type Ⅰ collagen gels with different stiffness and to investigate the effects of three-dimensional (3D) culture environments of the gels on the morphology, free migration ability, and cell killing function of natural killer (NK) cells. Methods Type Ⅰ collagen was isolated from the tails of Sprague Dawley (SD) rats and collagen gels with different levels of stiffnesses were prepared accordingly. The microstructure of the collagen gels was observed by laser confocal microscopy. The stiffness of the collagen gels was assessed by measuring the plateau modulus with a rheometer. NK-92MI cells were cultured in collagen gels with different levels of stiffness. The morphology of NK-92MI cells was observed by inverted microscope. High content imaging system was used to record the free migration process of NK-92MI cells and analyze the migration speed and distance. NK-92MI cells were cultured with type Ⅰ collagen gels with different levels of stiffness for 24 h and 48 h and, then, co-cultured with human colorectal DLD-1, a human adenocarcinoma epithelial cell line. CCK8 assay was performed to determine the proliferation rate of DLD-1 cells and analyze the cell killing ability of NK-92MI cells. Results Low-stiffness type Ⅰ collagen gel and high-stiffness type Ⅰ collagen gel with the respective stiffness of (10.970±2.10) Pa and (114.50±3.40) Pa were successfully prepared. Compared with those cultured with the low-stiffness type Ⅰ collagen gel, the NK-92MI cells in the high-stiffness type Ⅰ collagen gel showed a more elongated shape (P<0.05), the mean area of the cells was reduced ([69.88±26.97] μm2 vs. [46.59±21.62] μm2, P<0.05), the roundness of the cells decreased (0.82±0.12 vs. 0.78±0.18, P<0.05), cell migration speed decreased ([2.50±0.91] μm/min vs. [1.70±0.72] μm/min, P<0.001) and the migration distance was shortened ([147.10±53.74] μm vs. [98.03± 40.95] μm, P<0.0001), with all the differences being statistically significant. Compared with those cultured with the low-stiffness type Ⅰ collagen gel, NK-92MI cells cultured with high-stiffness type Ⅰ collagen gel for 24 h could promote DLD-1 cell proliferation, with the proliferation rate being (46.39±12.79)% vs. (65.87±4.45)% (P<0.05) and reduce the cell killing ability. Comparison of the cells cultured for 48 h led to similar results, with the proliferation rates being (31.36±2.88)% vs. (74.57±2.16)% (P<0.05), and the differences were all statistically significant. Conclusion The 3D culture environment of type Ⅰ collagen gels with different levels of stiffness alters the morphology, migration ability, and killing function of NK-92MI cells. This study provides the research basis for exploring and understanding the mechanisms by which the biomechanical microenvironment affects the immune response of NK cells, as well as laying the theoretical foundation for optimizing immunotherapy protocols.
Collapse
Affiliation(s)
- 世棋 龙
- 贵州医科大学基础医学院/生物与工程学院 免疫学教研室 (贵阳 550029)School of Basic Medicine/School of Biology and Engineering, Guizhou Medical University, Guiyang 550029, China
| | - 翠芳 吴
- 贵州医科大学基础医学院/生物与工程学院 免疫学教研室 (贵阳 550029)School of Basic Medicine/School of Biology and Engineering, Guizhou Medical University, Guiyang 550029, China
| | - 柱 曾
- 贵州医科大学基础医学院/生物与工程学院 免疫学教研室 (贵阳 550029)School of Basic Medicine/School of Biology and Engineering, Guizhou Medical University, Guiyang 550029, China
| |
Collapse
|
33
|
赵 川, 王 湘, 王 贵. [Hot Topics and Emerging Trends in Mechanobiology Research]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1-5. [PMID: 38322522 PMCID: PMC10839494 DOI: 10.12182/20240160104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Indexed: 02/08/2024]
Abstract
Mechanobiology focuses on a series of important physiopathological processes, such as how cells perceive different mechanomechanical stimuli, the process of intracellular mechanotransduction, and how mechanical signals determine the behavior and fate of cells. From the initial stage of embryogenesis, to developmental biology and regenerative medicine, or even through the whole life process, mechanical signaling cascades and cellular mechanical responses in mechanobiology are of great significance in biomedical research. In recent years, research in the field of mechanobiology has undergone remarkable development. Several scientific consortia around the world have been analyzing mechanobiological processes from different perspectives, aiming to gain insights into the regulatory mechanisms by which mechanical factors affect cell fate determination. In this article, we summarized and reviewed the topics that have attracted more research interests in recent years in the field of mechanobiology, for example, arterial blood vessels, stem cell, and ion channel. We also discussed the potential trends that may emerge, such as nuclear deformation, fibrous extracellular matrix, tumor mechanobiology, cellular mechanotransduction, and piezo ion channels. In addition, we put forward new ideas concerning the limitations of mechanism research and the importance of big data analysis and mining in this field, thereby providing objective support and a systematic framework for grasping the hot research topics and exploring new research directions in the field of mechanobiology.
Collapse
Affiliation(s)
- 川榕 赵
- 重庆大学生物工程学院,生物流变科学与技术教育部重点实验室,血管植入物开发国家地方联合工程实验室 (重庆 400045)College of Bioengineering, Chongqing University, Key Laboratory of Biorheology Science and Technology (Chongqing University), Ministry of Education, and State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400045, China
- 金凤实验室 (重庆 401329)JinFeng Laboratory, Chongqing 401329, China
| | - 湘秀 王
- 重庆大学生物工程学院,生物流变科学与技术教育部重点实验室,血管植入物开发国家地方联合工程实验室 (重庆 400045)College of Bioengineering, Chongqing University, Key Laboratory of Biorheology Science and Technology (Chongqing University), Ministry of Education, and State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400045, China
- 金凤实验室 (重庆 401329)JinFeng Laboratory, Chongqing 401329, China
| | - 贵学 王
- 重庆大学生物工程学院,生物流变科学与技术教育部重点实验室,血管植入物开发国家地方联合工程实验室 (重庆 400045)College of Bioengineering, Chongqing University, Key Laboratory of Biorheology Science and Technology (Chongqing University), Ministry of Education, and State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400045, China
- 金凤实验室 (重庆 401329)JinFeng Laboratory, Chongqing 401329, China
| |
Collapse
|
34
|
冯 唐, 杨 欣, 王 琦, 刘 肖. [Hepatocellular Carcinoma-Derived Exosomes: Key Players in Intercellular Communication Within the Tumor Microenvironment]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:6-12. [PMID: 38322525 PMCID: PMC10839483 DOI: 10.12182/20240160203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Indexed: 02/08/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths in the world. Due to the insidious onset and rapid progression and a lack of effective treatments, the prognosis of patients with HCC is extremely poor, with the average 5-year survival rate being less than 10%. The tumor microenvironment (TME), the internal environment in which HCC develops, can regulate the oncogenesis, development, invasion, and metastasis of HCC. During the process of cancer progression, HCC cells can regulate the biological behaviors of tumor cells, cancer-associated fibroblasts, cancer-associated immune cells, and other cells in the TME by releasing exosomes containing specific signals, thereby promoting cancer progression. However, the exact molecular mechanisms and the roles of exosomes in the specific cellular regulation of these processes are not fully understood. Herein, we summarized the TME components of HCC, the sources and the biological traits of exosomes in the TME, and the impact of mechanical factors on exosomes. In addition, special attention was given to the discussion of the effects of HCC-exosomes on different types of cells in the microenvironment. There are still many difficulties to be overcome before exosomes can be applied as carriers in clinical cancer treatment. First of all, the homogeneity of exosomes is difficult to ensure. Secondly, exosomes are mainly administered through subcutaneous injection. Although this method is simple and easy to implement, the absorption efficiency is not ideal. Thirdly, exosome extraction methods are limited in number and inefficient, making it difficult to prepare exosomes in large quantities. It is important to ensure that exosomes are used in sufficient quantities to trigger an effective tumor immune response, especially for exosome-mediated tumor immunotherapy. With the improvement in identification, isolation, and purification technology, exosomes are expected to be successfully used in the clinical diagnosis of early-stage HCC and the clinical treatment of liver cancer.
Collapse
Affiliation(s)
- 唐 冯
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 欣蕊 杨
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 琦为 王
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 肖珩 刘
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
35
|
Mierke CT. Extracellular Matrix Cues Regulate Mechanosensing and Mechanotransduction of Cancer Cells. Cells 2024; 13:96. [PMID: 38201302 PMCID: PMC10777970 DOI: 10.3390/cells13010096] [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: 11/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Extracellular biophysical properties have particular implications for a wide spectrum of cellular behaviors and functions, including growth, motility, differentiation, apoptosis, gene expression, cell-matrix and cell-cell adhesion, and signal transduction including mechanotransduction. Cells not only react to unambiguously mechanical cues from the extracellular matrix (ECM), but can occasionally manipulate the mechanical features of the matrix in parallel with biological characteristics, thus interfering with downstream matrix-based cues in both physiological and pathological processes. Bidirectional interactions between cells and (bio)materials in vitro can alter cell phenotype and mechanotransduction, as well as ECM structure, intentionally or unintentionally. Interactions between cell and matrix mechanics in vivo are of particular importance in a variety of diseases, including primarily cancer. Stiffness values between normal and cancerous tissue can range between 500 Pa (soft) and 48 kPa (stiff), respectively. Even the shear flow can increase from 0.1-1 dyn/cm2 (normal tissue) to 1-10 dyn/cm2 (cancerous tissue). There are currently many new areas of activity in tumor research on various biological length scales, which are highlighted in this review. Moreover, the complexity of interactions between ECM and cancer cells is reduced to common features of different tumors and the characteristics are highlighted to identify the main pathways of interaction. This all contributes to the standardization of mechanotransduction models and approaches, which, ultimately, increases the understanding of the complex interaction. Finally, both the in vitro and in vivo effects of this mechanics-biology pairing have key insights and implications for clinical practice in tumor treatment and, consequently, clinical translation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| |
Collapse
|
36
|
Swamy K. Therapeutic In Situ Cancer Vaccine Using Pulsed Stereotactic Body Radiotherapy-A Translational Model. Vaccines (Basel) 2023; 12:7. [PMID: 38276666 PMCID: PMC10819354 DOI: 10.3390/vaccines12010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Both radiation and cancer therapeutic vaccine research are more than 100 years old, and their potential is likely underexplored. Antiangiogenics, nanoparticle targeting, and immune modulators are some other established anticancer therapies. In the meantime, immunotherapy usage is gaining momentum in clinical applications. This article proposes the concept of a pulsed/intermittent/cyclical endothelial-sparing single-dose in situ vaccination (ISVRT) schedule distinguishable from the standard therapeutic stereotactic body radiotherapy (SBRT) and stereotactic radiosurgery (SRS) plans. This ISVRT schedule can repeatedly generate tumor-specific neoantigens and epitopes for primary and immune modulation effects, augment supplementary immune enhancement techniques, activate long-term memory cells, avoid extracellular matrix fibrosis, and essentially synchronize with the vascular normalized immunity cycle. The core mechanisms of ISVRT impacting in situ vaccination would be optimizing cascading antigenicity and adjuvanticity. The present proposed hypothesis can be validated using the algorithm presented. The indications for the proposed concept are locally progressing/metastatic cancers that have failed standard therapies. Immunotherapy/targeted therapy, chemotherapy, antiangiogenics, and vascular-lymphatic normalization are integral to such an approach.
Collapse
|
37
|
Horta CA, Doan K, Yang J. Mechanotransduction pathways in regulating epithelial-mesenchymal plasticity. Curr Opin Cell Biol 2023; 85:102245. [PMID: 37804773 PMCID: PMC10796216 DOI: 10.1016/j.ceb.2023.102245] [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: 07/10/2023] [Revised: 08/16/2023] [Accepted: 09/03/2023] [Indexed: 10/09/2023]
Abstract
The extracellular matrix (ECM) provides structural support for cells and mediates cell-stromal communications. In addition to ECM proteins, mechanical force exerted from the ECM serves as a critical regulator of many biological processes. Epithelial-mesenchymal transition (EMT) is a cellular process by which epithelial cells loosen their cellular junctions and migrate and invade in a more mesenchymal fashion. Recent studies show that increasing ECM stiffness can impinge on cellular signaling pathways through mechanotransduction to promote carcinoma cells to undergo EMT, suggesting that mechanical force exerted by the ECM plays a critical role in tumor invasion and metastasis. Here, we highlight recent work utilizing innovative approaches to study mechanotransduction and summarize newly discovered mechanisms by which mechanosensors and responders regulate EMT during tumor progression and metastasis.
Collapse
Affiliation(s)
- Calista A Horta
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Khoa Doan
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
38
|
Parodi I, Di Lisa D, Pastorino L, Scaglione S, Fato MM. 3D Bioprinting as a Powerful Technique for Recreating the Tumor Microenvironment. Gels 2023; 9:482. [PMID: 37367152 DOI: 10.3390/gels9060482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023] Open
Abstract
In vitro three-dimensional models aim to reduce and replace animal testing and establish new tools for oncology research and the development and testing of new anticancer therapies. Among the various techniques to produce more complex and realistic cancer models is bioprinting, which allows the realization of spatially controlled hydrogel-based scaffolds, easily incorporating different types of cells in order to recreate the crosstalk between cancer and stromal components. Bioprinting exhibits other advantages, such as the production of large constructs, the repeatability and high resolution of the process, as well as the possibility of vascularization of the models through different approaches. Moreover, bioprinting allows the incorporation of multiple biomaterials and the creation of gradient structures to mimic the heterogeneity of the tumor microenvironment. The aim of this review is to report the main strategies and biomaterials used in cancer bioprinting. Moreover, the review discusses several bioprinted models of the most diffused and/or malignant tumors, highlighting the importance of this technique in establishing reliable biomimetic tissues aimed at improving disease biology understanding and high-throughput drug screening.
Collapse
Affiliation(s)
- Ilaria Parodi
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
- National Research Council of Italy, Institute of Electronic, Computer and Telecommunications Engineering (IEIIT), 16149 Genoa, Italy
| | - Donatella Di Lisa
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
| | - Laura Pastorino
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
| | - Silvia Scaglione
- National Research Council of Italy, Institute of Electronic, Computer and Telecommunications Engineering (IEIIT), 16149 Genoa, Italy
- React4life S.p.A., 16152 Genova, Italy
| | - Marco Massimo Fato
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
- National Research Council of Italy, Institute of Electronic, Computer and Telecommunications Engineering (IEIIT), 16149 Genoa, Italy
| |
Collapse
|
39
|
Papavassiliou KA, Basdra EK, Papavassiliou AG. The emerging promise of tumour mechanobiology in cancer treatment. Eur J Cancer 2023; 190:112938. [PMID: 37390803 DOI: 10.1016/j.ejca.2023.112938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/05/2023] [Indexed: 07/02/2023]
Abstract
Tumour cell biomechanics has lately came to the fore as a disparate feature that fosters cancer development and progression. Tumour mechanosensing entails a mechanical interplay amongst tumour cells, extracellular matrix (ECM) and cells of the tumour microenvironment (TME). Sensory receptors (mechanoceptors) detect changes of extracellular mechanical inputs such as various types of mechanical forces/stress and trigger oncogenic signalling pathways advocating for cancer initiation, growth, survival, angiogenesis, invasion, metastasis, and immune evasion. Moreover, alterations in ECM stiffness and potentiation of mechanostimulated transcriptional regulatory molecules (transcription factors/cofactors) have been shown to strongly correlate with resistance to anticancer drugs. On this basis, new mechanosensitive proteins emerge as potential therapeutic targets and/or biomarkers in cancer. Accordingly, tumour mechanobiology arises as a promising field that can potentially provide novel combinatorial regimens to reverse drug resistance, as well as offer unprecedented targeting approaches that may help to more effectively treat a large proportion of solid tumours and their complications. Here, we highlight recent findings regarding various aspects of tumour mechanobiology in the clinical setting and discuss evidence-based perspectives of developing diagnostic/prognostic tools and therapeutic approaches that exploit tumour-TME physical associations.
Collapse
Affiliation(s)
- Kostas A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthimia K Basdra
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
40
|
Hsia CR, Melters DP, Dalal Y. The Force is Strong with This Epigenome: Chromatin Structure and Mechanobiology. J Mol Biol 2023; 435:168019. [PMID: 37330288 PMCID: PMC10567996 DOI: 10.1016/j.jmb.2023.168019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 06/19/2023]
Abstract
All life forms sense and respond to mechanical stimuli. Throughout evolution, organisms develop diverse mechanosensing and mechanotransduction pathways, leading to fast and sustained mechanoresponses. Memory and plasticity characteristics of mechanoresponses are thought to be stored in the form of epigenetic modifications, including chromatin structure alterations. These mechanoresponses in the chromatin context share conserved principles across species, such as lateral inhibition during organogenesis and development. However, it remains unclear how mechanotransduction mechanisms alter chromatin structure for specific cellular functions, and if altered chromatin structure can mechanically affect the environment. In this review, we discuss how chromatin structure is altered by environmental forces via an outside-in pathway for cellular functions, and the emerging concept of how chromatin structure alterations can mechanically affect nuclear, cellular, and extracellular environments. This bidirectional mechanical feedback between chromatin of the cell and the environment can potentially have important physiological implications, such as in centromeric chromatin regulation of mechanobiology in mitosis, or in tumor-stroma interactions. Finally, we highlight the current challenges and open questions in the field and provide perspectives for future research.
Collapse
Affiliation(s)
- Chieh-Ren Hsia
- Chromatin Structure and Epigenetic Mechanisms, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, NCI, NIH, Bethesda, MD, United States. https://twitter.com/JeremiahHsia
| | - Daniël P Melters
- Chromatin Structure and Epigenetic Mechanisms, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, NCI, NIH, Bethesda, MD, United States. https://twitter.com/dpmelters
| | - Yamini Dalal
- Chromatin Structure and Epigenetic Mechanisms, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, NCI, NIH, Bethesda, MD, United States. https://twitter.com/NCIYaminiDalal
| |
Collapse
|
41
|
Portale F, Di Mitri D. NK Cells in Cancer: Mechanisms of Dysfunction and Therapeutic Potential. Int J Mol Sci 2023; 24:ijms24119521. [PMID: 37298470 DOI: 10.3390/ijms24119521] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Natural killer cells (NK) are innate lymphocytes endowed with the ability to recognize and kill cancer cells. Consequently, adoptive transfer of autologous or allogeneic NK cells represents a novel opportunity in cancer treatment that is currently under clinical investigation. However, cancer renders NK cells dysfunctional, thus restraining the efficacy of cell therapies. Importantly, extensive effort has been employed to investigate the mechanisms that restrain NK cell anti-tumor function, and the results have offered forthcoming solutions to improve the efficiency of NK cell-based therapies. The present review will introduce the origin and features of NK cells, summarize the mechanisms of action and causes of dysfunction of NK cells in cancer, and frame NK cells in the tumoral microenvironment and in the context of immunotherapies. Finally, we will discuss therapeutic potential and current limitations of NK cell adoptive transfer in tumors.
Collapse
Affiliation(s)
- Federica Portale
- Tumor Microenviroment Unit, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Diletta Di Mitri
- Tumor Microenviroment Unit, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
| |
Collapse
|
42
|
Hansen E, Holaska JM. The nuclear envelope and metastasis. Oncotarget 2023; 14:317-320. [PMID: 37057891 PMCID: PMC10103595 DOI: 10.18632/oncotarget.28375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Indexed: 04/15/2023] Open
Affiliation(s)
| | - James M. Holaska
- Correspondence to:James M. Holaska, Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; Rowan University Graduate School of Biomedical Sciences, Stratford, NJ 08084, USA email
| |
Collapse
|
43
|
Purushothaman A, Mohajeri M, Lele TP. The role of glycans in the mechanobiology of cancer. J Biol Chem 2023; 299:102935. [PMID: 36693448 PMCID: PMC9930169 DOI: 10.1016/j.jbc.2023.102935] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Although cancer is a genetic disease, physical changes such as stiffening of the extracellular matrix also commonly occur in cancer. Cancer cells sense and respond to extracellular matrix stiffening through the process of mechanotransduction. Cancer cell mechanotransduction can enhance cancer-promoting cell behaviors such as survival signaling, proliferation, and migration. Glycans, carbohydrate-based polymers, have recently emerged as important mediators and/or modulators of cancer cell mechanotransduction. Stiffer tumors are characterized by increased glycan content on cancer cells and their associated extracellular matrix. Here we review the role of cancer-associated glycans in coupled mechanical and biochemical alterations during cancer progression. We discuss the recent evidence on how increased expression of different glycans, in the form of glycoproteins and proteoglycans, contributes to both mechanical changes in tumors and corresponding cancer cell responses. We conclude with a summary of emerging tools that can be used to modify glycans for future studies in cancer mechanobiology.
Collapse
Affiliation(s)
- Anurag Purushothaman
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA.
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Tanmay P Lele
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA; Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA; Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA; Department of Translational Medical Sciences, Texas A&M University, Houston, Texas, USA.
| |
Collapse
|
44
|
Zhou Y, Cheng L, Liu L, Li X. NK cells are never alone: crosstalk and communication in tumour microenvironments. Mol Cancer 2023; 22:34. [PMID: 36797782 PMCID: PMC9933398 DOI: 10.1186/s12943-023-01737-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Immune escape is a hallmark of cancer. The dynamic and heterogeneous tumour microenvironment (TME) causes insufficient infiltration and poor efficacy of natural killer (NK) cell-based immunotherapy, which becomes a key factor triggering tumour progression. Understanding the crosstalk between NK cells and the TME provides new insights for optimising NK cell-based immunotherapy. Here, we present new advances in direct or indirect crosstalk between NK cells and 9 specialised TMEs, including immune, metabolic, innervated niche, mechanical, and microbial microenvironments, summarise TME-mediated mechanisms of NK cell function inhibition, and highlight potential targeted therapies for NK-TME crosstalk. Importantly, we discuss novel strategies to overcome the inhibitory TME and provide an attractive outlook for the future.
Collapse
Affiliation(s)
- Yongqiang Zhou
- grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000 China ,grid.412643.60000 0004 1757 2902Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China ,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Lu Cheng
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Lu Liu
- grid.412643.60000 0004 1757 2902Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China. .,Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China. .,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China.
| |
Collapse
|
45
|
Cancer-Associated Adipocytes and Breast Cancer: Intertwining in the Tumor Microenvironment and Challenges for Cancer Therapy. Cancers (Basel) 2023; 15:cancers15030726. [PMID: 36765683 PMCID: PMC9913307 DOI: 10.3390/cancers15030726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Adipocytes are the main components in breast tissue, and cancer-associated adipocytes (CAAs) are one of the most important components in the tumor microenvironment of breast cancer (BC). Bidirectional regulation was found between CAAs and BC cells. BC facilitates the dedifferentiation of adjacent adipocytes to form CAAs with morphological and biological changes. CAAs increase the secretion of multiple cytokines and adipokines to promote the tumorigenesis, progression, and metastasis of BC by remodeling the extracellular matrix, changing aromatase expression, and metabolic reprogramming, and shaping the tumor immune microenvironment. CAAs are also associated with the therapeutic response of BC and provide potential targets in BC therapy. The present review provides a comprehensive description of the crosstalk between CAAs and BC and discusses the potential strategies to target CAAs to overcome BC treatment resistance.
Collapse
|
46
|
Zhang J, Wang Y, Yuan B, Qin H, Wang Y, Yu H, Teng X, Yang Y, Zou J, Zhang M, Huang W, Wang Y. Identifying key transcription factors and immune infiltration in non-small-cell lung cancer using weighted correlation network and Cox regression analyses. Front Oncol 2023; 13:1112020. [PMID: 37197420 PMCID: PMC10183566 DOI: 10.3389/fonc.2023.1112020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/12/2023] [Indexed: 05/19/2023] Open
Abstract
Introduction Lung cancer is one of the most common cancers and a significant cause of cancer-related deaths. Non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases. Therefore, it is crucial to identify effective diagnostic and therapeutic methods. In addition, transcription factors are essential for eukaryotic cells to regulate their gene expression, and aberrant expression transcription factors are an important step in the process of oncogenesis in NSCLC. Methods Differentially expressed transcription factors between NSCLC and normal tissues by analyzing mRNA profiling from The Cancer Genome Atlas (TCGA) database program were identified. Weighted correlation network analysis (WGCNA) and line plot of least absolute shrinkage and selection operator (LASSO) were performed to find prognosis-related transcription factors. The cellular functions of transcription factors were performed by 5-ethynyl-2'-deoxyuridine (EdU) assay, wound healing assay, cell invasion assay in lung cancer cells. Results We identified 725 differentially expressed transcription factors between NSCLC and normal tissues. Three highly related modules for survival were discovered, and transcription factors highly associated with survival were obtained by using WGCNA. Then line plot of LASSO was applied to screen transcription factors related to prognosis and build a prognostic model. Consequently, SETDB2, SNAI3, SCML4, and ZNF540 were identified as prognosis-related transcription factors and validated in multiple databases. The low expression of these hub genes in NSCLC was associated with poor prognosis. The deletions of both SETDB2 and SNAI3 were found to promote proliferation, invasion, and stemness in lung cancer cells. Furthermore, there were significant differences in the proportions of 22 immune cells between the high- and low-score groups. Discussion Therefore, our study identified the transcription factors involved in regulating NSCLC, and we constructed a panel for the prediction of prognosis and immune infiltration to inform the clinical application of transcription factor analysis in the prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Jingyao Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinuo Wang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baowen Yuan
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Qin
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Wang
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hefen Yu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xu Teng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yunkai Yang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zou
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Huang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- *Correspondence: Wei Huang, ; Yan Wang,
| | - Yan Wang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Wei Huang, ; Yan Wang,
| |
Collapse
|
47
|
Chen X, Xia Q, Sun N, Zhou H, Xu Z, Yang X, Yan R, Li P, Li T, Qin X, Yang H, Wu C, You F, Liao X, Li S, Liu Y. Shear stress enhances anoikis resistance of cancer cells through ROS and NO suppressed degeneration of Caveolin-1. Free Radic Biol Med 2022; 193:95-107. [PMID: 36243211 DOI: 10.1016/j.freeradbiomed.2022.10.271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/08/2022] [Accepted: 10/08/2022] [Indexed: 12/13/2022]
Abstract
Circulating tumor cells (CTCs) acquire enhanced anti-anoikis abilities after experiencing flow shear stress in the circulatory system. Our previous study demonstrated that low shear stress (LSS) promotes anoikis resistance of human breast carcinoma cells via caveolin-1 (Cav-1)-dependent extrinsic and intrinsic apoptotic pathways. However, the underlying mechanism how LSS enhanced Cav-1 expression in suspended cancer cells remains unclear. Herein, we found that LSS induced redox signaling was involved in the regulation of Cav-1 level and anoikis resistance in suspension cultured cancer cells. Exposure of human breast carcinoma MDA-MB-231 cells to LSS (2 dyn/cm2) markedly induced ROS and •NO generation, which promoted the cell viability and reduced the cancer cell apoptosis. Furthermore, ROS and •NO scavenging inhibited the upregulation of Cav-1 by interfering ubiquitination, and suppressed the anoikis resistance of suspended tumor cells. These findings provide new insight into the mechanism by which LSS-stimulated ROS and •NO generation increases Cav-1 stabilization in suspended cancer cells through inhibition of ubiquitination and proteasomal degradation, which could be a potential target for therapy of metastatic tumors.
Collapse
Affiliation(s)
- Xiangyan Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Qiong Xia
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Ningwei Sun
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Hailei Zhou
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Zhihao Xu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Xi Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Ran Yan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Ping Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Tingting Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Xiang Qin
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Hong Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Chunhui Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, PR China
| | - Shun Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China.
| | - Yiyao Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China.
| |
Collapse
|