1
|
Yi L, Xie H, Zhang X, Gu M, Zhang K, Xia T, Pan S, Yin H, Wu R, You Y, You B. LPAR3 and COL8A1, as matrix stiffness-related biomarkers, promote nasopharyngeal carcinoma metastasis by triggering EMT and angiogenesis. Cell Signal 2025; 131:111712. [PMID: 40049264 DOI: 10.1016/j.cellsig.2025.111712] [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: 12/02/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Matrix stiffness affects the progression of nasopharyngeal carcinoma, but the underlying mechanism is still unknown. Here, we demonstrated that nasopharyngeal carcinoma tissues with distant metastasis contain large collagen deposits and strong matrix stiffness. First, we performed RNA-seq analysis of nasopharyngeal carcinoma cells cultured on polyacrylamide hydrogel systems and found that LPAR3 and COL8A1 are potential matrix stiffness markers. Based on in vivo and in vitro experiments, matrix stiffness mainly affected tumor metastasis rather than proliferation. Subsequently, we found that matrix stiffness triggers the formation of epithelial-mesenchymal transition by increasing the expression of LPAR3 in nasopharyngeal carcinoma, which is related to metastasis. In addition, matrix stiffness promotes the expression of COL8A1 secreted by nasopharyngeal carcinoma and is related to tumor angiogenesis. Simultaneous inhibition of LPAR3 and COL8A1 genes significantly reduced nasopharyngeal carcinoma invasion and metastasis. Based on the investigation, we confirmed that matrix stiffness governs the progression of nasopharyngeal carcinoma and that LPAR3 and COL8A1, as matrix stiffness related biomarkers, promote nasopharyngeal carcinoma metastasis by inducing epithelial-mesenchymal transition and angiogenesis. Overall, the in-depth exploration of matrix stiffness may provide a strategy for clinical treatment intervention and provide promising targets for clinical nasopharyngeal carcinoma treatment.
Collapse
MESH Headings
- Humans
- Epithelial-Mesenchymal Transition
- Nasopharyngeal Carcinoma/pathology
- Nasopharyngeal Carcinoma/metabolism
- Nasopharyngeal Carcinoma/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/genetics
- Receptors, Lysophosphatidic Acid/metabolism
- Receptors, Lysophosphatidic Acid/genetics
- Nasopharyngeal Neoplasms/pathology
- Nasopharyngeal Neoplasms/metabolism
- Nasopharyngeal Neoplasms/genetics
- Cell Line, Tumor
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Animals
- Extracellular Matrix/metabolism
- Neoplasm Metastasis
- Mice, Nude
- Mice
- Gene Expression Regulation, Neoplastic
- Mice, Inbred BALB C
- Cell Proliferation
- Female
- Male
- Angiogenesis
Collapse
Affiliation(s)
- Lu Yi
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Nantong University, Qixiu Road 19, Nantong 226001, Jiangsu Province, China
| | - Haijing Xie
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China
| | - Xin Zhang
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Nantong University, Qixiu Road 19, Nantong 226001, Jiangsu Province, China
| | - Miao Gu
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China
| | - Kaiwen Zhang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China
| | - Tian Xia
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China
| | - Si Pan
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China
| | - Haimeng Yin
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Nantong University, Qixiu Road 19, Nantong 226001, Jiangsu Province, China
| | - Rui Wu
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Nantong University, Qixiu Road 19, Nantong 226001, Jiangsu Province, China
| | - Yiwen You
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China.
| | - Bo You
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
2
|
Ahn W, Han J, Kim N, Hwang YH, Kim W, Lee Y, Lee DY, Cheong IW, Han K, Nam GH, Kim IS, Lee EJ. Hierarchical protein nano-crystalline hydrogel with extracellular vesicles for ectopic lymphoid structure formation. Biomaterials 2025; 318:123166. [PMID: 39933315 DOI: 10.1016/j.biomaterials.2025.123166] [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: 10/17/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Among cancer therapies, immune checkpoint blockade (ICB) has emerged as a prominent approach, substantially enhancing anti-tumor immune responses. However, the efficacy of ICB is often limited in the absence of a pre-existing immune response within the tumor microenvironment. Here, we introduce a novel hierarchical protein hydrogel platform designed to facilitate the formation of artificial tertiary lymphoid structures (aTLS), thereby improving ICB efficacy. Through the integration of self-assembling ferritin protein nanocages, rec1-resilin protein, and CP05 peptide, our hierarchical hydrogels provide a structurally supportive and functionally adaptive scaffold capable of on-demand self-repair in response to mild thermal treatments. The effective encapsulation of extracellular vesicles (EVs) via the CP05 peptide ensures the formation of aTLS with germinal center-like structures within the hierarchical hydrogel. We demonstrate that, combined with ICB therapy, EV-loaded hierarchical hydrogels also induce the TLS within the tumor, markedly promoting immune responses against ICB-resistant tumor. This bioactive hydrogel platform offers a versatile tool for enhancing a broad range of immunotherapies, with potential applications extending beyond TLS to other frameworks that support complex tissue architectures.
Collapse
Affiliation(s)
- Wonkyung Ahn
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea; Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jihoon Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Nayeon Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeong Ha Hwang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Wonjun Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeram Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dong Yun Lee
- Department of Polymer Science and Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - In Woo Cheong
- Department of Applied Chemistry, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Koohee Han
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Gi-Hoon Nam
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
3
|
Xi J, Ji C, Sun H, Wu Y, Shi C, Li S, Yang T, Shen Y, Li Y, Fan Y, Zhao Q, Liu S, Xie T, Chen G. Research progress on new physical therapies for cancer (Review). Oncol Lett 2025; 29:313. [PMID: 40337606 PMCID: PMC12056479 DOI: 10.3892/ol.2025.15059] [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: 12/28/2024] [Accepted: 04/04/2025] [Indexed: 05/09/2025] Open
Abstract
Currently, the clinical treatment of cancer is mainly based on surgery, chemotherapy and radiotherapy, but there are still problems associated with these treatments, such as disease recurrence and adverse reactions. The complexity and harmful nature of cancer mean that combining multiple treatment methods is an inevitable response. Therefore, it is of theoretical and practical significance to expand upon and study the aforementioned classic and traditional measures. With the advancement of technology, physical therapy has become important in the current research and treatment of cancer, and the physical factors related to cancer deserve in-depth study and discussion. The present review aimed to describe the mechanisms of action of pressure, temperature, photo-, sound and other physical therapies for cancer, which may provide new avenues for cancer treatment.
Collapse
Affiliation(s)
- Jingyi Xi
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Cheng Ji
- Department of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Haixin Sun
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yurun Wu
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Chengjie Shi
- Department of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Shasha Li
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Tao Yang
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yuxiang Shen
- Department of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yulin Li
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yaoxuan Fan
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Qichao Zhao
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Shuiping Liu
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Tian Xie
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Gongxing Chen
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| |
Collapse
|
4
|
Horisaka H, Yokawa S, Suzuki R, Emoto R, Maeda R, Furuno T. Suppression of FcεRI-evoked Degranulation in RBL-2H3 Cells on Gelatin Methacryloyl Hydrogel. Cell Biochem Biophys 2025; 83:2481-2488. [PMID: 39731647 DOI: 10.1007/s12013-024-01657-3] [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] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
Cell-extracellular matrix (ECM) interactions play multiple roles in developmental, physiological, and pathological processes. ECM stiffness substantially affects cellular morphology, migration, and function. In this study, we investigated the effect of ECM comprising gelatin methacryloyl (GelMA) on the activation of rat basophilic leukemia (RBL-2H3) cells, a model mast cell line. Maintenance of intracellular Ca2+ concentration ([Ca2+]i) elevation and subsequent degranulation, evoked by crosslinking the high-affinity IgE receptors (FcεRI), were significantly suppressed in RBL-2H3 cells on collagen-coated GelMA hydrogel than those on collagen-coated glass dishes and plastic wells. Thapsigargin and phorbol myristate acetate caused sustained [Ca2+]i increase and degranulation to a similar extent in cells on both GelMA hydrogel and plastic wells/glass dishes. F-actin was clearly accumulated along the periphery of RBL-2H3 cells in plane attached to glass, but not GelMA hydrogel, suggesting that the loose actin cytoskeleton of RBL-2H3 cells on GelMA hydrogel caused suppressive degranulation through unstable FcεRI aggregation.
Collapse
Affiliation(s)
- Haruna Horisaka
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Satoru Yokawa
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Ruriko Suzuki
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Rin Emoto
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Rino Maeda
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Tadahide Furuno
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| |
Collapse
|
5
|
Li X, Zhao H, Jiang E, Liu P, Chen Y, Wang Y, Li J, Wu Y, Liu Z, Shang Z. ITGB1/FERMT1 mechanoactivation enhances CD44 characteristic stemness in oral squamous cell carcinoma via ubiquitin-dependent CK1α degradation. Oncogene 2025; 44:1530-1544. [PMID: 40044983 DOI: 10.1038/s41388-025-03317-z] [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: 08/20/2024] [Revised: 01/30/2025] [Accepted: 02/18/2025] [Indexed: 05/15/2025]
Abstract
Cancer stem cells (CSCs) contribute to chemotherapy resistance and poor prognosis, posing significant challenges in the treatment of oral squamous cell carcinoma. The extracellular matrix (ECM)-constructed microenvironment remodels the niche of CSCs. Yet mechanisms by which biophysical properties of ECM relate to CSCs remain undefined. Here, our findings link ECM mechanical stimuli to CSCs phenotype transition, and propose that ECM stiffening mechanoactivates tumor cells to dedifferentiate and acquire CD44+ stem cell-like characteristics through noncanonical mechanotransduction. ITGB1 senses and transduces biomechanical signals, while FERMT1 acts as an intracellular mechanotransduction downstream, activating CSCs. Mechanistically, FERMT1 promotes the proteasomal degradation of CK1α by E3 ubiquitin ligase MIB1, thereby triggering Wnt signaling pathway. Combining targeted ECM softening with mechanotransduction inhibition strategy significantly attenuates tumor stemness and chemoresistance in vivo. Therefore, our findings highlight the role of ECM in regulating CSCs via biomechanical-dependent manner, suggesting the ECM/ITGB1/FERMT1/Wnt axis as a promising therapeutic target for CSCs therapy.
Collapse
Affiliation(s)
- Xiang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Erhui Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Pan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ji Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yufei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhenan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Bull EC, Singh A, Harden AM, Soanes K, Habash H, Toracchio L, Carrabotta M, Schreck C, Shah KM, Riestra PV, Chantoiseau M, Da Costa MEM, Moquin-Beaudry G, Pantziarka P, Essiet EA, Gerrand C, Gartland A, Bojmar L, Fahlgren A, Marchais A, Papakonstantinou E, Tomazou EM, Surdez D, Heymann D, Cidre-Aranaz F, Fromigue O, Sexton DW, Herold N, Grünewald TGP, Scotlandi K, Nathrath M, Green D. Targeting metastasis in paediatric bone sarcomas. Mol Cancer 2025; 24:153. [PMID: 40442778 PMCID: PMC12121159 DOI: 10.1186/s12943-025-02365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/22/2025] [Indexed: 06/02/2025] Open
Abstract
Paediatric bone sarcomas (e.g. Ewing sarcoma, osteosarcoma) comprise significant biological and clinical heterogeneity. This extreme heterogeneity affects response to systemic therapy, facilitates inherent and acquired drug resistance and possibly underpins the origins of metastatic disease, a key component implicit in cancer related death. Across all cancers, metastatic models have offered competing accounts on when dissemination occurs, either early or late during tumorigenesis, whether metastases at different foci arise independently and directly from the primary tumour or give rise to each other, i.e. metastases-to-metastases dissemination, and whether cell exchange occurs between synchronously growing lesions. Although it is probable that all the above mechanisms can lead to metastatic disease, clinical observations indicate that distinct modes of metastasis might predominate in different cancers. Around 70% of patients with bone sarcoma experience metastasis during their disease course but the fundamental molecular and cell mechanisms underlying spread are equivocal. Newer therapies such as tyrosine kinase inhibitors have shown promise in reducing metastatic relapse in trials, nonetheless, not all patients respond and 5-year overall survival remains at ~ 50%. Better understanding of potential bone sarcoma biological subgroups, the role of the tumour immune microenvironment, factors that promote metastasis and clinical biomarkers of prognosis and drug response are required to make progress. In this review, we provide a comprehensive overview of the approaches to manage paediatric patients with metastatic Ewing sarcoma and osteosarcoma. We describe the molecular basis of the tumour immune microenvironment, cell plasticity, circulating tumour cells and the development of the pre-metastatic niche, all required for successful distant colonisation. Finally, we discuss ongoing and upcoming patient clinical trials, biomarkers and gene regulatory networks amenable to the development of anti-metastasis medicines.
Collapse
Affiliation(s)
- Emma C Bull
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Archana Singh
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
- Amity Institute of Biotechnology, Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurugram, India
| | - Amy M Harden
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Kirsty Soanes
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Hala Habash
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Lisa Toracchio
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Marianna Carrabotta
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Christina Schreck
- Children's Cancer Research Center, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Karan M Shah
- School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Paulina Velasco Riestra
- Biomedical and Clinical Sciences, Division of Surgery, Orthopaedics and Oncology, Linköping University, Linköping, Sweden
| | | | - Maria Eugénia Marques Da Costa
- Gustave Roussy Institute, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Pan Pantziarka
- Anticancer Fund, Meise, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | | | - Craig Gerrand
- Orthopaedic Oncology, Royal National Orthopaedic Hospital, Stanmore, UK
| | - Alison Gartland
- School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Linda Bojmar
- Biomedical and Clinical Sciences, Division of Surgery, Orthopaedics and Oncology, Linköping University, Linköping, Sweden
| | - Anna Fahlgren
- Biomedical and Clinical Sciences, Division of Cell and Neurobiology, Linköping University, Linköping, Sweden
| | | | - Evgenia Papakonstantinou
- Pediatric Hematology-Oncology, Ippokratio General Hospital of Thessaloniki, Thessaloniki, Greece
| | - Eleni M Tomazou
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Didier Surdez
- Faculty of Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Dominique Heymann
- School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
- UMR6286, Nantes Université, CNRS, US2B, Nantes, France
- Institut de Cancérologie de L'Ouest, Saint-Herblain, France
| | - Florencia Cidre-Aranaz
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Translational Pediatric Sarcoma Research, German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Olivia Fromigue
- Inserm UMR981, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Darren W Sexton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Nikolas Herold
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Paediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas G P Grünewald
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Translational Pediatric Sarcoma Research, German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Michaela Nathrath
- Children's Cancer Research Center, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Pediatric Oncology, Klinikum Kassel, Kassel, Germany
| | - Darrell Green
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK.
| |
Collapse
|
7
|
Guo Y, Ye J, Shemesh A, Odeh A, Trebicz-Geffen M, Wolfenson H, Ankri S. Enteropathogenic Escherichia coli induces Entamoeba histolytica superdiffusion movement on fibronectin by reducing traction forces. PLoS Pathog 2025; 21:e1012618. [PMID: 40408453 DOI: 10.1371/journal.ppat.1012618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 05/02/2025] [Indexed: 05/25/2025] Open
Abstract
Amebiasis, caused by Entamoeba histolytica, is a global health concern, affecting millions and causing significant mortality, particularly in areas with poor sanitation. Although recent studies have examined E. histolytica's interaction with human intestinal microbes, the impact of bacterial presence on the parasite's motility, mechanical forces, and their potential role in altering invasiveness have not been fully elucidated. In this study, we utilized a micropillar-array system combined with live imaging to investigate the effects of enteropathogenic Escherichia coli on E. histolytica's motility characteristics, F-actin spatial localization, and traction force exerted on fibronectin-coated substrates. Our findings indicate that co-incubation with live enteropathogenic E. coli significantly enhances the motility of E. histolytica, as evidenced by superdiffusive movement-characterized by increased directionality and speed-resulting in broader dispersal and more extensive tissue/cell damage. This increased motility is accompanied by a reduction in F-actin-dependent traction forces and podosome-like structures on fibronectin-coated substrates, but with increased F-actin localization in the upper part of the cytoplasm. These findings highlight the role of physical interactions and cellular behaviors in modulating the parasite's virulence, providing new insights into the mechanistic basis of its pathogenicity.
Collapse
Affiliation(s)
- Yuanning Guo
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jun Ye
- Department of Molecular Microbiology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ariel Shemesh
- Biomedical core facilities, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Anas Odeh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Meirav Trebicz-Geffen
- Department of Molecular Microbiology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Serge Ankri
- Department of Molecular Microbiology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
8
|
Shah S, D'Souza GGM. Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis. Cells 2025; 14:732. [PMID: 40422235 DOI: 10.3390/cells14100732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor-stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models.
Collapse
Affiliation(s)
- Shrey Shah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
- Atom Bioworks Inc., Cary, NC 27513, USA
| | - Gerard G M D'Souza
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| |
Collapse
|
9
|
Kulkarni S, Tebar F, Rentero C, Zhao M, Sáez P. Competing signaling pathways controls electrotaxis. iScience 2025; 28:112329. [PMID: 40292314 PMCID: PMC12032939 DOI: 10.1016/j.isci.2025.112329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/01/2024] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Understanding how cells follow exogenous cues is a key question for biology, medicine, and bioengineering. Growing evidence shows that electric fields represent a precise and programmable method to control cell migration. Most data suggest that the polarization of membrane proteins and the following downstream signaling are central to electrotaxis. Unfortunately, how these multiple mechanisms coordinate with the motile machinery of the cell is still poorly understood. Here, we develop a mechanistic model that explains electrotaxis across different cell types. Using the zebrafish proteome, we identify membrane proteins directly related to migration signaling pathways that polarize anodally and cathodally. Further, we show that the simultaneous and asymmetric distribution of these membrane receptors establish multiple cooperative and competing stimuli for directing the anodal and cathodal migration of the cell. Using electric fields, we enhance, cancel, or switch directed cell migration, with clear implications in promoting tissue regeneration or arresting tumor progression.
Collapse
Affiliation(s)
- S. Kulkarni
- Laboratori de Càlcul Numèric (LaCàN), ETS de Ingeniería de Caminos, Canales y Puertos, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - F. Tebar
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Cell Compartments and Signaling Group, Fundació de Recerca Clínic Barcelona - Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - C. Rentero
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Cell Compartments and Signaling Group, Fundació de Recerca Clínic Barcelona - Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - M. Zhao
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - P. Sáez
- Laboratori de Càlcul Numèric (LaCàN), ETS de Ingeniería de Caminos, Canales y Puertos, Universitat Politècnica de Catalunya, Barcelona, Spain
- IMTech (Institute of Mathematics), Universitat Politècnica de Catalunya-BarcelonaTech., 08034 Barcelona, Spain
| |
Collapse
|
10
|
Galanti A, Rosetti B, Valente S, Braidotti N, Sbacchi M, Todros S, Pavan P, Gobbo P. A Photonastic Prototissue Capable of Photo-Mechano-Chemical Transduction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2502830. [PMID: 40350988 DOI: 10.1002/adma.202502830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/07/2025] [Indexed: 05/14/2025]
Abstract
Despite recent significant advances in the controlled assembly of protocell units into complex 3D architectures, the development of prototissues capable of mimicking the sophisticated energy transduction processes fundamental to living tissues remains a critical unmet challenge in bottom-up synthetic biology. Here a synthetic approach is described to start addressing this challenge and report the bottom-up chemical construction of a photonastic prototissue endowed with photo-mechano-chemical transduction capabilities. For this, novel protocells enclosing photothermal transducing proto-organelles based on gold nanoparticles and a thermoresponsive polymeric proto-cortex are developed. These advanced protocell units are assembled into prototissues capable of light-induced reversible contractions and complex motions, which can be exploited to reversibly switch off a coordinated internalized enzyme metabolism by blocking the access of small substrate molecules. Overall, the work provides a synthetic pathway to constructing prototissues with sophisticated energy transduction mechanisms, enabling the rational design of emergent behaviors in synthetic materials and addressing critical challenges in bottom-up synthetic biology and bioinspired materials engineering.
Collapse
Affiliation(s)
- Agostino Galanti
- Department of Chemical and Pharmaceutical Sciences, Università degli Studi di Trieste, Via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Beatrice Rosetti
- Department of Chemical and Pharmaceutical Sciences, Università degli Studi di Trieste, Via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Stefano Valente
- Department of Chemical and Pharmaceutical Sciences, Università degli Studi di Trieste, Via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Nicoletta Braidotti
- Department of Chemical and Pharmaceutical Sciences, Università degli Studi di Trieste, Via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Maria Sbacchi
- Department of Chemical and Pharmaceutical Sciences, Università degli Studi di Trieste, Via L. Giorgieri, 1, Trieste, 34127, Italy
- National Interuniversity Consortium of Materials Science and Technology, Unit of Trieste, Via G. Giusti 9, Firenze, 50121, Italy
| | - Silvia Todros
- Department of Industrial Engineering, Università degli Studi di Padova, Padova, 35131, Italy
| | - Piero Pavan
- Department of Industrial Engineering, Università degli Studi di Padova, Padova, 35131, Italy
- Tissue Engineering Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti, 4F, Padova, 35127, Italy
| | - Pierangelo Gobbo
- Department of Chemical and Pharmaceutical Sciences, Università degli Studi di Trieste, Via L. Giorgieri, 1, Trieste, 34127, Italy
- National Interuniversity Consortium of Materials Science and Technology, Unit of Trieste, Via G. Giusti 9, Firenze, 50121, Italy
| |
Collapse
|
11
|
Liu S, Cao H, Wang Z, Zhu J, An X, Zhang L, Song Y. Single-cell transcriptomics reveals extracellular matrix remodeling and collagen dynamics during lactation in sheep mammary gland. Int J Biol Macromol 2025; 312:143669. [PMID: 40319976 DOI: 10.1016/j.ijbiomac.2025.143669] [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: 12/03/2024] [Revised: 04/13/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The mammary gland is a dynamic organ with diverse cell populations that maintain glandular homeostasis, particularly during lactation. However, the cellular architecture and molecular mechanisms underlying lactational remodeling in the sheep mammary gland remain incompletely understood. Given similarities in mammary stromal structure, sheep serve as a valuable model for studying lactational changes relevant to the human breast, which experiences collagen loss and sagging during lactation. Utilizing single-cell transcriptomics (scRNA-seq), we mapped the sheep mammary gland's cellular landscape at postpartum days 60 and 150, identifying seven major cell types, including six distinct epithelial clusters. These clusters revealed differentiation among luminal progenitors, hormone-sensing, and myoepithelial cells across peak and late lactation stages. Transcriptomic analysis highlighted pivotal roles for epithelial integrity and ECM remodeling, with myoepithelial cells centrally involved in these processes. We observed significant collagen remodeling driven by fibroblast-epithelial crosstalk and ECM reorganization during late lactation. Comparative analysis with human mammary epithelial cells showed conserved basal and myoepithelial cell populations, while luminal cells diverged across species. This study provides insights into lactation biology and ECM remodeling, offering a framework to inform future studies on lactational adaptation and its implications for human health.
Collapse
Affiliation(s)
- Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhanhang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Junru Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
12
|
McMahon AMB, Twigg MS, Marchant R, Banat IM. The Application of Glycolipid-Type Microbial Biosurfactants as Active Pharmaceutical Ingredients for the Treatment and Prevention of Cancer. Pharmaceuticals (Basel) 2025; 18:676. [PMID: 40430495 PMCID: PMC12115046 DOI: 10.3390/ph18050676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Pharmaceutical scientists have researched the potential of secondary metabolites biosynthesized by microorganisms as active pharmaceutical ingredients (APIs) for the treatment of cancer. Ideally, these APIs should possess anticancer bioactivity that specifically targets tumor cells while having little cytotoxic effect on healthy tissue. Biosurfactants are microbial secondary metabolites with surface-active properties and individual bioactivities that have the potential to either destroy cancer cells in a targeted fashion or prevent tumor cell formation. Currently, the best-studied class of microbial biosurfactants for the purpose of anticancer bioactivity is glycolipids, which contain a hydrophilic sugar moiety bonded to a hydrophobic fatty acid. Anticancer investigations are mainly carried out using in vitro models that show that compounds belonging to each of the four sub-classes of microbial glycolipid have significant anticancer bioactivity. The targeted action of this activity appears to be highly dependent on a specific congener molecular structure with nuanced alterations in structure leading to the killing of both tumor and healthy cells. This review compiles the current literature relating to glycolipid anticancer activity and provides a critical appraisal of exploiting the bioactivity of these compounds as novel anticancer agents. Finally, we propose several suggestions on how this research could be improved moving forward via method standardization.
Collapse
Affiliation(s)
| | | | | | - Ibrahim M. Banat
- Pharmaceutical Science Research Group, Biomedical Science Research Institute, Ulster University, Coleraine BT52 1SA, UK; (A.M.B.M.); (M.S.T.); (R.M.)
| |
Collapse
|
13
|
Ciccone G, Azevedo Gonzalez‐Oliva M, Versaevel M, Cantini M, Vassalli M, Salmeron‐Sanchez M, Gabriele S. Epithelial Cell Mechanoresponse to Matrix Viscoelasticity and Confinement Within Micropatterned Viscoelastic Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408635. [PMID: 39950757 PMCID: PMC12079340 DOI: 10.1002/advs.202408635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/21/2025] [Indexed: 05/16/2025]
Abstract
Extracellular matrix (ECM) viscoelasticity has emerged as a potent regulator of physiological and pathological processes, including cancer progression. Spatial confinement within the ECM is also known to influence cell behavior in these contexts. However, the interplay between matrix viscoelasticity and spatial confinement in driving epithelial cell mechanotransduction is not well understood, as it relies on experiments employing purely elastic hydrogels. This work presents an innovative approach to fabricate and micropattern viscoelastic polyacrylamide hydrogels with independently tuneable Young's modulus and stress relaxation, specifically designed to mimic the mechanical properties observed during breast tumor progression, transitioning from a soft dissipative tissue to a stiff elastic one. Using this platform, this work demonstrates that matrix viscoelasticity differentially modulates breast epithelial cell spreading, adhesion, YAP nuclear import and cell migration, depending on the initial stiffness of the matrix. Furthermore, by imposing spatial confinement through micropatterning, this work demonstrates that confinement alters cellular responses to viscoelasticity, including cell spreading, mechanotransduction and migration. These findings establish ECM viscoelasticity as a key regulator of epithelial cell mechanoresponse and highlight the critical role of spatial confinement in soft, dissipative ECMs, which was a previously unexplored aspect.
Collapse
Affiliation(s)
- Giuseppe Ciccone
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Mechanobiology & Biomaterials GroupUniversity of MonsResearch Institute for BiosciencesCIRMAP, Place du ParcMons20 B‐7000Belgium
- Centre for the Cellular MicroenvironmentUniversity of GlasgowAdvanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
| | - Mariana Azevedo Gonzalez‐Oliva
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Centre for the Cellular MicroenvironmentUniversity of GlasgowAdvanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
| | - Marie Versaevel
- Mechanobiology & Biomaterials GroupUniversity of MonsResearch Institute for BiosciencesCIRMAP, Place du ParcMons20 B‐7000Belgium
| | - Marco Cantini
- Centre for the Cellular MicroenvironmentUniversity of GlasgowAdvanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
| | - Massimo Vassalli
- Centre for the Cellular MicroenvironmentUniversity of GlasgowAdvanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
| | - Manuel Salmeron‐Sanchez
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Centre for the Cellular MicroenvironmentUniversity of GlasgowAdvanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Sylvain Gabriele
- Mechanobiology & Biomaterials GroupUniversity of MonsResearch Institute for BiosciencesCIRMAP, Place du ParcMons20 B‐7000Belgium
| |
Collapse
|
14
|
Suzuki M, Kawauchi K, Machiyama H, Hirata H, Ishiwata S, Fujita H. Dynamic Remodeling of Mechano-Sensing Complexes in Suspended Fibroblast Cell-Sheets Under External Mechanical Stimulus. Biotechnol Bioeng 2025. [PMID: 40270085 DOI: 10.1002/bit.28996] [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: 11/29/2024] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025]
Abstract
Freestanding cell-sheets are valuable bio-materials for use in regenerative medicine and tissue engineering. Because cell-sheets experience various mechanical stimulations during handling, it is important to understand the responses of cells to these stimulations. Here, we demonstrate changes in the localization of various proteins during the stretching of fibroblast cell-sheets. These proteins are known to be involved in mechano-sensing. Upon stretching, actin filaments appear parallel to the stretching direction. At cell-cell junctions, β-catenin forms clusters that co-localize with accumulated vinculin and zyxin as well as the actin filaments. The p130 Crk-associated substrate, known to be present in focal adhesions, is also recruited to these clusters and phosphorylated. Our results suggest that mechano-sensing machinery is formed at cell-cell junctions when the cell-sheets are stretched.
Collapse
Affiliation(s)
- Madoka Suzuki
- Institute for Protein Research, The University of Osaka, Suita, Osaka, Japan
| | - Keiko Kawauchi
- Faculty of Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo, Japan
| | - Hiroaki Machiyama
- Department of Immunology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hiroaki Hirata
- Department of Life Science and Biotechnology, Kanazawa Institute of Technology, Hakusan, Ishikawa, Japan
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Hideaki Fujita
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Hiroshima, Japan
| |
Collapse
|
15
|
Surjadi JU, Aymon BFG, Carton M, Portela CM. Double-network-inspired mechanical metamaterials. NATURE MATERIALS 2025:10.1038/s41563-025-02219-5. [PMID: 40269148 DOI: 10.1038/s41563-025-02219-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 03/19/2025] [Indexed: 04/25/2025]
Abstract
Mechanical metamaterials can achieve high stiffness and strength at low densities, but often at the expense of low ductility and stretchability-a persistent trade-off in materials. In contrast, double-network hydrogels feature interpenetrating compliant and stiff polymer networks, and exhibit unprecedented combinations of high stiffness and stretchability, resulting in exceptional toughness. Here we present double-network-inspired metamaterials by integrating monolithic truss (stiff) and woven (compliant) components into a metamaterial architecture, which achieves a tenfold increase in stiffness and stretchability compared to its pure counterparts. Nonlinear computational mechanics models elucidate that enhanced energy dissipation in these double-network-inspired metamaterials stems from increased frictional dissipation due to entanglements between networks. Through introduction of internal defects, which typically degrade mechanical properties, we demonstrate a threefold increase in energy dissipation for these metamaterials via failure delocalization. This work opens avenues for developing metamaterials in a high-compliance regime inspired by polymer network topologies.
Collapse
Affiliation(s)
- James Utama Surjadi
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bastien F G Aymon
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Molly Carton
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, University of Maryland, College Park, MD, USA
| | - Carlos M Portela
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
16
|
Depierre M, Mularski A, Ruppel A, Le Clainche C, Balland M, Niedergang F. A crosstalk between adhesion and phagocytosis integrates macrophage functions into their microenvironment. iScience 2025; 28:112067. [PMID: 40177633 PMCID: PMC11964680 DOI: 10.1016/j.isci.2025.112067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/25/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Phagocytosis is the process of actin-dependent internalization and degradation of large particles. Macrophages, which are professional phagocytes, are present in all tissues and are, thus, exposed to environments with different mechanical properties. How mechanical cues from macrophages' environment affect their ability to phagocytose and, in turn, how phagocytosis influences how phagocytic cells interact with their environment remain poorly understood. We found that the ability of macrophages to perform phagocytosis varied with the substrate stiffness. Using live traction force microscopy, we showed that phagocytosing macrophages applied more dynamic traction forces to their substrate. In addition, integrin-mediated phagocytosis triggered a transient loss of podosomes that was associated with decreased degradation of the extracellular matrix, concomitantly with RhoA activation and F-actin recruitment at phagocytic cups. Overall, these results highlight a crosstalk between macrophage phagocytosis and cell adhesion. Mechanical properties of the microenvironment influence phagocytosis, which, in turn, impacts how macrophages interact with their surroundings.
Collapse
Affiliation(s)
- Manon Depierre
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| | - Anna Mularski
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| | - Artur Ruppel
- Université Grenoble Alpes, CNRS, Interdisciplinary Laboratory of Physics (LIPhy), Grenoble, France
| | - Christophe Le Clainche
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Martial Balland
- Université Grenoble Alpes, CNRS, Interdisciplinary Laboratory of Physics (LIPhy), Grenoble, France
| | | |
Collapse
|
17
|
von Eysmondt H, Seifert J, Rheinlaender J, Schäffer TE. Mechanosensing alters platelet migration. Acta Biomater 2025; 196:213-221. [PMID: 39986637 DOI: 10.1016/j.actbio.2025.02.042] [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: 10/04/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 02/24/2025]
Abstract
Platelets have long been established as a safeguard of our vascular system. Recently, haptotactic platelet migration has been discovered as a part of the immune response. In addition, platelets exhibit mechanosensing properties, changing their behavior in response to the stiffness of the underlying substrate. However, the influence of substrate stiffness on platelet migration behavior remains elusive. Here, we investigated the migration of platelets on fibrinogen-coated polydimethylsiloxane (PDMS) substrates with different stiffnesses. Using phase-contrast and fluorescence microscopy as well as a deep-learning neural network, we tracked single migrating platelets and measured their migration distance and velocity. We found that platelets migrated on stiff PDMS substrates (E = 2 MPa), while they did not migrate on soft PDMS substrates (E = 5 kPa). Platelets migrated also on PDMS substrates with intermediate stiffness (E = 100 kPa), but their velocity and the fraction of migrating platelets were diminished compared to platelets on stiff PDMS substrates. The straightness of platelet migration, however, was not significantly influenced by substrate stiffness. We used scanning ion conductance microscopy (SICM) to image the three-dimensional shape of migrating platelets, finding that platelets on soft substrates did not show the polarization and shape change associated with migration. Furthermore, the fibrinogen density gradient, which is generated by migrating platelets, was reduced for platelets on soft substrates. Our work demonstrates that substrate stiffness, and thus platelet mechanosensing, influences platelet migration. Substrate stiffness for optimal platelet migration is quite high (>100 kPa) in comparison to other cell types, with possible implications on platelet behavior in inflammatory and injured tissue. STATEMENT OF SIGNIFICANCE: Platelets can feel and react to the stiffness of their surroundings - a process called mechanosensation. Additionally, platelets migrate via substrate-bound fibrinogen as part of the innate immune response during injury or inflammation. It has been shown that the migration of immune cells is influenced by the stiffness of the underlying substrate, but the effect of substrate stiffness on the migration of platelets has not yet been investigated. Using differently stiff substrates made from PDMS, we show that substrate stiffness affects platelet migration. Stiff substrates facilitate fast and frequent platelet migration with a strong platelet shape anisotropy and a strong fibrinogen removal while soft substrates inhibit platelet migration. These findings highlight the influence of the stiffness of the surrounding tissue on the platelet immune response, possibly enhancing platelet migration in inflamed tissue.
Collapse
Affiliation(s)
| | - Jan Seifert
- Institute of Applied Physics, University of Tübingen, Germany
| | | | | |
Collapse
|
18
|
Field EH, Ratcliffe J, Johnson CJ, Binger KJ, Reynolds NP. Self-healing, 3D printed bioinks from self-assembled peptide and alginate hybrid hydrogels. BIOMATERIALS ADVANCES 2025; 169:214145. [PMID: 39675342 DOI: 10.1016/j.bioadv.2024.214145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/17/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
There is a pressing need for new cell-laden, printable, biomaterials that are rigid and highly biocompatible. These materials can mimic stiffer tissues such as cartilage, fibrotic tissue and cancer microenvironments, and thus have exciting applications in regenerative medicine, wound healing and cancer research. Self-assembled peptides (SAPs) functionalised with aromatic groups such as Fluorenyl-9-methoxycarbonyl (Fmoc) show promise as components of these biomaterials. However, the harsh basic conditions often used to solubilise SAPs leads to issues with toxicity and reproducibility. Here, we have designed a hybrid material comprised of self-assembled Fmoc-diphenylalanine (Fmoc-FF) assemblies dispersed throughout a sodium alginate matrix and investigated the influence of different organic solvents as peptide solubilising agents. Bioinks fabricated from peptides dissolved in 1,1,1,3,3,3-Hexafluoro-2-propanol (HFIP) showed improved biocompatibility compared to those made from Dimethyl Sulfoxide (DMSO) peptide stocks, due to the increased volatility and reduced surface tension of HFIP, allowing for more efficient expulsion from the system. Through optimisation of assembly and solvent conditions we can generate hybrid bioinks with stiffnesses up to 8 times greater than sodium alginate alone that remain highly printable, even when laden with high concentrations of cells. In addition, the shear-thinning nature of the self-assembled peptide assemblies gave the hybrid bioinks highly desirable self-healing capabilities. Our developed hybrid materials allow the bioprinting of materials previously considered too stiff to extrude without causing shear induced cytotoxicity with applications in tissue engineering and biosensing.
Collapse
Affiliation(s)
- Emily H Field
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Julian Ratcliffe
- La Trobe University Bioimaging platform, La Trobe University, Australia, Melbourne, Victoria 3086, Australia
| | - Chad J Johnson
- La Trobe University Bioimaging platform, La Trobe University, Australia, Melbourne, Victoria 3086, Australia
| | - Katrina J Binger
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia; Centre for Cardiovascular Biology & Disease Research, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia
| | - Nicholas P Reynolds
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia; The Biomedical and Environmental Sensor Technology (BEST) Research Centre, Biosensors Program, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
19
|
Quan T, Qin Z, He T, Fisher GJ. Integrin α11β1 as a Key Collagen Receptor in Human Skin Dermis: Insight into Fibroblast Function and Skin Dermal Aging. J Invest Dermatol 2025:S0022-202X(25)00364-1. [PMID: 40139564 DOI: 10.1016/j.jid.2025.03.006] [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: 11/21/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025]
Abstract
Collagen-binding integrins play a crucial role in facilitating fibroblast-collagen interactions and regulating cellular functions. In this study, we identified that among 4 collagen-binding integrins, integrin α11 was the predominant type in human skin dermal fibroblasts and that loss of integrin α11 expression contributed to skin dermal aging. Integrin α11β1 was critical for regulating fibroblast-collagen interactions, including cell adhesion, spreading, morphology, mechanical tension, and the production of collagenous extracellular matrix. TGF-β was recognized as the primary regulator of integrin α11 expression. Notably, dermal fibroblasts in aged human skin demonstrated impaired TGF-β signaling, which coincided with a loss of integrin α11 expression, whereas the expression of other collagen-binding integrins remained unchanged. Similarly, in senescent dermal fibroblasts in vitro, impaired TGF-β signaling was associated with a significant reduction in integrin α11 expression, whereas other collagen-binding integrins were upregulated or unaffected. Furthermore, collapsed dermal fibroblasts, a key characteristic of dermal fibroblasts in aged human skin, specifically downregulated integrin α11, whereas other collagen-binding integrins were upregulated or remained unchanged. These findings suggest a negative feedback loop in which an impaired TGF-β-integrin α11β1 axis and fibroblast collapse promote dermal aging in human skin. This self-reinforcing cycle reflects the progressive and unidirectional nature of biological aging.
Collapse
Affiliation(s)
- Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | - Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tianyuan He
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gary J Fisher
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
20
|
Ishihara S, Enomoto A, Sakai A, Iida T, Tange S, Kioka N, Nukuda A, Nagasato AI, Yasuda M, Tokino T, Haga H. Stiff extracellular matrix activates the transcription factor ATF5 to promote the proliferation of cancer cells. iScience 2025; 28:112057. [PMID: 40124511 PMCID: PMC11928855 DOI: 10.1016/j.isci.2025.112057] [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: 01/04/2024] [Revised: 07/20/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer tissues are stiffer than normal tissues. Carcinogenesis stiffens the extracellular matrix (ECM) of cancerous tissues, to which cancer cells respond by activating transcription factors, such as YAP/TAZ, Twist1, and β-catenin, which further elevate their malignancy. However, these transcription factors are also expressed in normal tissues. Therefore, inhibiting these factors in order to treat cancer may lead to severe side effects. Here, we show that activating transcription factor 5 (ATF5), highly expressed in tumors, is activated by ECM stiffness and promotes the proliferation of cancer cells, including that of pancreatic cancer cells and lung cancer cells. In addition, ATF5 suppressed the expression of early growth response 1 (EGR1), thereby accelerating cancer cell proliferation. Stiff ECMs trigger the JAK-MYC pathway which activates ATF5. JAK activation was actomyosin independent whereas MYC induction was actomyosin dependent. These results demonstrate the critical role played by ATF5 in the mechanotransduction process seen in cancers.
Collapse
Affiliation(s)
- Seiichiro Ishihara
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Akihiro Sakai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550 Japan
| | - Shoichiro Tange
- Department of Medical Genome Sciences, Cancer Research Institute, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Akihiro Nukuda
- Transdisciplinary Life Science Course, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810 Japan
| | - Ayaka Ichikawa Nagasato
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Motoaki Yasuda
- Department of Oral Molecular Microbiology, Division of Oral Pathobiological Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060-8586, Japan
| | - Takashi Tokino
- Department of Medical Genome Sciences, Cancer Research Institute, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Hisashi Haga
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| |
Collapse
|
21
|
Clemens C, Gehring R, Riedl P, Pompe T. Matrix deformation and mechanotransduction as markers of breast cancer cell phenotype alteration at matrix interfaces. Biomater Sci 2025; 13:1578-1589. [PMID: 39960148 DOI: 10.1039/d4bm01589d] [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: 03/12/2025]
Abstract
The dissemination of metastatic cells from the primary tumor into the surrounding tissue is a key event in the progression of cancer. This process involves the migration of cells across defined tissue interfaces that separate the dense tumor tissue from the adjacent healthy tissue. Prior research showed that cell transmigration across collagen I matrix interfaces induces a switch towards a more aggressive phenotype including a change in directionality of migration and chemosensitivity correlated to increased DNA damage during transmigration. Hence, mechanical forces acting at the nucleus during transmigration are hypothesized to trigger phenotype switching. Here, we present results from a particle image velocimetry (PIV) based live cell analysis of breast cancer cell transmigration across sharp matrix interfaces constituted of two collagen type I networks with different pore sizes. We found strong and highly localized collagen network deformation caused by cellular forces at the moment of crossing interfaces from dense into open matrices. Additionally, an increased contractility of transmigrated cells was determined for cells with the switch phenotype. Moreover, studies on mechanotransductive signaling at the nucleus, emerin translocation and YAP activation, indicated a misregulation of these signals for transmigrated cells with altered phenotype. These findings show that matrix interfaces between networks of different pore sizes mechanically challenge invasive breast cancer cells during transmigration by a strong asymmetry of contracting forces, impeding nuclear mechanotransduction pathways, with a subsequent trigger of more aggressive phenotypes.
Collapse
Affiliation(s)
- Cornelia Clemens
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Rosa Gehring
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Philipp Riedl
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Tilo Pompe
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| |
Collapse
|
22
|
Moradi Kashkooli F, Mirala F, H H Tehrani M, Alirahimi M, Souri M, Golzaryan A, Kar S, Soltani M. Mechanical Forces in Tumor Growth and Treatment: Perspectives From Biology, Physics, Engineering, and Mathematical Modeling. WIREs Mech Dis 2025; 17:e70000. [PMID: 40170456 DOI: 10.1002/wsbm.70000] [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: 07/21/2023] [Revised: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 04/03/2025]
Abstract
The progression of tumors is influenced by mechanical forces and biological elements, such as hypoxia and angiogenesis. Mechanical factors, including stress, pressure, interstitial fluid pressure, and cellular traction forces, compromise normal tissue architecture, augmenting stiffness and thus promoting tumor growth and invasion. The selective elimination of specific tumor components can reduce growth-induced mechanical stress, thereby improving therapeutic efficacy. Furthermore, stress-relief drugs have the potential in enhancing chemotherapy outcomes. In this setting, computational modeling functions as an essential tool for quantitatively elucidating the mechanical principles underlying tumor formation. These models can precisely replicate the impact of mechanical pressures on solid tumors, offering insight into the regulation of tumor behavior by these forces. Tumor growth produces mechanical forces, including compression, displacement, and deformation, leading to irregular stress patterns, expedited tumor advancement, and reduced treatment efficacy. This review analyzes the impact of mechanical forces on carcinogenesis and solid tumor proliferation, emphasizing the significance of stress alleviation in regulating tumor growth. Furthermore, we investigate the influence of mechanical forces on tumor dissemination and emphasize the promise of integrating computational modeling with force-targeted cancer therapies to improve treatment efficacy by tackling the fundamental mechanics of tumor proliferation.
Collapse
Affiliation(s)
| | - Fatemeh Mirala
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Masoud H H Tehrani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mahvash Alirahimi
- Department of Obstetrics & Gynecology, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Aryan Golzaryan
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Egaila, Kuwait
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
23
|
Handschin C, Shalhoub H, Mazet A, Guyon C, Dusserre N, Boutet-Robinet E, Oliveira H, Guillermet-Guibert J. Biotechnological advances in 3D modeling of cancer initiation. Examples from pancreatic cancer research and beyond. Biofabrication 2025; 17:022008. [PMID: 40018875 DOI: 10.1088/1758-5090/adb51c] [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: 07/19/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
In recent years, biofabrication technologies have garnered significant attention within the scientific community for their potential to create advancedin vitrocancer models. While these technologies have been predominantly applied to model advanced stages of cancer, there exists a pressing need to develop pertinent, reproducible, and sensitive 3D models that mimic cancer initiation lesions within their native tissue microenvironment. Such models hold profound relevance for comprehending the intricacies of cancer initiation, to devise novel strategies for early intervention, and/or to conduct sophisticated toxicology assessments of putative carcinogens. Here, we will explain the pivotal factors that must be faithfully recapitulated when constructing these models, with a specific focus on early pancreatic cancer lesions. By synthesizing the current state of research in this field, we will provide insights into recent advances and breakthroughs. Additionally, we will delineate the key technological and biological challenges that necessitate resolution in future endeavors, thereby paving the way for more accurate and insightfulin vitrocancer initiation models.
Collapse
Affiliation(s)
- C Handschin
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - H Shalhoub
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
| | - A Mazet
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - C Guyon
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - N Dusserre
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - E Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - H Oliveira
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - J Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| |
Collapse
|
24
|
Ranjan P, Devi C, Verma N, Bansal R, Srivastava VK, Das P. Understanding the Role of MicroRNAs in Congenital Tooth Agenesis: A Multi-omics Integration. Biochem Genet 2025:10.1007/s10528-025-11064-9. [PMID: 39985697 DOI: 10.1007/s10528-025-11064-9] [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: 11/20/2024] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
This study employs a comprehensive multi-omics approach to investigate the regulatory roles of specific microRNAs (miRNAs) in Congenital Tooth Agenesis (CTA). A total of 58 miRNAs associated with tooth diseases, cancer, and bone development were initially identified through a literature review and analyzed using bioinformatics. Based on target prediction and network analysis, eight miRNAs with strong connectivity and common target genes were shortlisted for further investigation. Blood samples from 10 CTA patients and 5 healthy controls were analyzed for miRNA expression using stem-loop RT-PCR. Four miRNAs-hsa-miR-218-5p, hsa-miR-15b-5p, hsa-miR-200b-3p, and hsa-let-7a-3p-were identified as significantly differentially expressed, marking their first reported involvement in CTA. Notably, hsa-miR-218-5p and hsa-let-7a-3p emerged as novel regulators with no prior associations with CTA or tooth development. To address the limitations of a small sample size, a multi-omics strategy was employed to validate these findings, integrating miRNA expression data with whole exome sequencing (WES), gene expression panels, and metabolomic profiling. The analysis confirmed the association of these four miRNAs with CTA and highlighted their involvement in critical biological pathways such as Wnt signaling, FGF signaling, and PI3 kinase pathways, which are essential for cellular proliferation, differentiation, and tissue morphogenesis. Importantly, the identification of these miRNAs in blood samples, rather than traditional dental tissues, highlights a minimally invasive approach that could aid in the early detection, therapeutic targeting, and personalized management of dental anomalies.
Collapse
Affiliation(s)
- Prashant Ranjan
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Chandra Devi
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Neha Verma
- Dentistry Oral Surgery and Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, 221005, India
- Dr Bhimrao Ramji Ambedkar Government Medical College, Kannauj, UP, India
| | - Rajesh Bansal
- Dentistry Oral Surgery and Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Vinay Kumar Srivastava
- Dentistry Oral Surgery and Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Parimal Das
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India.
| |
Collapse
|
25
|
Segovia C, Desrosiers V, Khadangi F, Robitaille K, Armero VS, D'Astous M, Khelifi G, Bergeron A, Hussein S, Richer M, Bossé Y, Fradet Y, Fradet V, Bilodeau S. A versatile and efficient method to isolate nuclei from low-input cryopreserved tissues for single-nuclei transcriptomics. Sci Rep 2025; 15:5581. [PMID: 39955438 PMCID: PMC11829965 DOI: 10.1038/s41598-025-90070-8] [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: 11/12/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
Clinical samples are vital for understanding diseases, but their scarcity requires refined research methods. Emerging single-cell technologies offer detailed views of tissue heterogeneity but need sufficient fully characterized tissues. We developed an optimized single-nuclei RNA sequencing (snRNA-seq) protocol to extract nuclei from just 15 mg of cryopreserved human tissue. Applied to four cancer tissues (brain, bladder, lung, prostate), it profiled 1550-7468 nuclei per tissue, revealing heterogeneity comparable to public single-cell atlases. This method enhances the use and sharing of rare, cryopreserved biospecimens, supporting research where sample quantity is limited and full tissue characterization is needed.
Collapse
Affiliation(s)
- Cristopher Segovia
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
| | - Vincent Desrosiers
- Centre de recherche du CHU de Québec - Université Laval, Axe Maladies Infectieuses Et Immunitaires, Québec, Québec, G1V 4G2, Canada
- Centre de recherche ARThrite de L'Université Laval, Québec, Québec, G1V 4G2, Canada
| | - Fatemeh Khadangi
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
| | - Karine Robitaille
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
- Centre de recherche NUTRISS - Nutrition, Santé Et Société - de L'Université Laval, Québec, Québec, G1V 4G2, Canada
- Institut sur la nutrition et les aliments fonctionnels de l'Université Laval, Québec, Québec, G1V 4G2, Canada
| | - Victoria Saavedra Armero
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, Québec, G1V 4G5, Canada
| | - Myreille D'Astous
- CHU de Québec - Université Laval, Québec, Québec, G1R 2J6, Canada
- Centre de recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, Québec, G1V 4G2, Canada
| | - Gabriel Khelifi
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
| | - Alain Bergeron
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, Québec, G1V 0A6, Canada
| | - Samer Hussein
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
- Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Québec, G1V 0A6, Canada
| | - Maxime Richer
- CHU de Québec - Université Laval, Québec, Québec, G1R 2J6, Canada
- Centre de recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, Québec, G1V 4G2, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, Québec, G1V 4G5, Canada
- Département de médecine moléculaire, Faculté de Médecine, Université Laval, Québec, Québec, G1V 0A6, Canada
| | - Yves Fradet
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
- CHU de Québec - Université Laval, Québec, Québec, G1R 2J6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, Québec, G1V 0A6, Canada
| | - Vincent Fradet
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada
- Centre de recherche NUTRISS - Nutrition, Santé Et Société - de L'Université Laval, Québec, Québec, G1V 4G2, Canada
- Institut sur la nutrition et les aliments fonctionnels de l'Université Laval, Québec, Québec, G1V 4G2, Canada
- CHU de Québec - Université Laval, Québec, Québec, G1R 2J6, Canada
| | - Steve Bilodeau
- Centre de recherche du CHU de Québec - Université Laval, Axe Oncologie, 1401, 18e rue, Québec, Québec, G1J 1Z4, Canada.
- Centre de recherche sur le cancer de l'Université Laval, Québec, Québec, G1R 3S3, Canada.
- Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Québec, G1V 0A6, Canada.
- Centre de recherche en données massives de l'Université Laval, Québec, Québec, G1V 0A6, Canada.
| |
Collapse
|
26
|
Little MJ, Mason JM, Mehrban N. Evolution of branched peptides as novel biomaterials. J Mater Chem B 2025; 13:2226-2241. [PMID: 39835399 PMCID: PMC11747965 DOI: 10.1039/d4tb01897d] [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: 08/22/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Branched peptide-based materials draw inspiration from dendritic structures to emulate the complex architecture of native tissues, aiming to enhance the performance of biomaterials in medical applications. These innovative materials benefit from several key features: they exhibit slower degradation rates, greater stiffness, and the ability to self-assemble. These properties are crucial for maintaining the structural integrity and functionality of the materials over time. By integrating bioactive peptides and natural polymers within their branched frameworks, these materials offer modularity and tunability and can accommodate a range of mechanical properties, degradation rates, and biological functions making them suitable for biomedical applications, including drug delivery systems, wound healing scaffolds, and tissue engineering constructs. In drug delivery, these materials can be engineered to release therapeutic agents in a controlled manner, enhancing the efficacy and safety of treatments. In wound healing, they provide a supportive environment which promotes rapid and efficient tissue repair. The combination of biomimetic design and functional adaptability makes branched peptide-based materials a promising candidate for the development of next-generation biomaterials, paving the way for significant advancements in healthcare.
Collapse
Affiliation(s)
| | - Jody M Mason
- University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | - Nazia Mehrban
- University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
27
|
Sharip A, Kunz J. Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis Progression and Therapy. Cells 2025; 14:266. [PMID: 39996739 PMCID: PMC11854242 DOI: 10.3390/cells14040266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Liver fibrosis, a consequence of chronic liver injury, represents a major global health burden and is the leading cause of liver failure, morbidity, and mortality. The pathological hallmark of this condition is excessive extracellular matrix deposition, driven primarily by integrin-mediated mechanotransduction. Integrins, transmembrane heterodimeric proteins that serve as primary ECM receptors, orchestrate complex mechanosignaling networks that regulate the activation, differentiation, and proliferation of hepatic stellate cells and other ECM-secreting myofibroblasts. These mechanical signals create self-reinforcing feedback loops that perpetuate the fibrotic response. Recent advances have provided insight into the roles of specific integrin subtypes in liver fibrosis and revealed their regulation of key downstream effectors-including transforming growth factor beta, focal adhesion kinase, RhoA/Rho-associated, coiled-coil containing protein kinase, and the mechanosensitive Hippo pathway. Understanding these mechanotransduction networks has opened new therapeutic possibilities through pharmacological manipulation of integrin-dependent signaling.
Collapse
Affiliation(s)
- Aigul Sharip
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
- Laboratory of Bioinformatics and Systems Biology, National Laboratory Astana, Astana 020000, Kazakhstan
| | - Jeannette Kunz
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
| |
Collapse
|
28
|
Agrawal A, Javanmardi Y, Watson SA, Serwinski B, Djordjevic B, Li W, Aref AR, Jenkins RW, Moeendarbary E. Mechanical signatures in cancer metastasis. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:3. [PMID: 39917412 PMCID: PMC11794153 DOI: 10.1038/s44341-024-00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/20/2024] [Indexed: 02/09/2025]
Abstract
The cancer metastatic cascade includes a series of mechanical barrier-crossing events, involving the physical movement of cancer cells from their primary location to a distant organ. This review describes the physical changes that influence tumour proliferation, progression, and metastasis. We identify potential mechanical signatures at every step of the metastatic cascade and discuss some latest mechanobiology-based therapeutic interventions to highlight the importance of interdisciplinary approaches in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ayushi Agrawal
- Department of Mechanical Engineering, University College London, London, UK
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, UK
| | - Sara A. Watson
- Department of Mechanical Engineering, University College London, London, UK
- Division of Biosciences, University College London, London, UK
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, UK
- Northeastern University London, London, UK
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, UK
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Amir R. Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Russell W. Jenkins
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| |
Collapse
|
29
|
Saez P, Shirke PU, Seth JR, Alegre-Cebollada J, Majumder A. Competing elastic and viscous gradients determine directional cell migration. Math Biosci 2025; 380:109362. [PMID: 39701208 DOI: 10.1016/j.mbs.2024.109362] [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: 06/16/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024]
Abstract
Cell migration regulates central life processes including embryonic development, tissue regeneration, and tumor invasion. To establish the direction of migration, cells follow exogenous cues. Durotaxis, the directed cell migration towards elastic stiffness gradients, is the classical example of mechanical taxis. However, whether gradients of the relaxation properties in the extracellular matrix may also induce tactic responses (viscotaxis) is not well understood. Moreover, whether and how durotaxis and viscotaxis interact with each other has never been investigated. Here, we integrate clutch models for cell adhesions with an active gel theory of cell migration to reveal the mechanisms that govern viscotaxis. We show that viscotaxis is enabled by an asymmetric expression of cell adhesions that further polarize the intracellular motility forces to establish the cell front, similar to durotaxis. More importantly, when both relaxation and elastic gradients coexist, durotaxis appears more efficient in controlling directed cell migration, which we confirm with experimental results. However, the presence of opposing relaxation gradients to an elastic one can arrest or shift the migration direction. Our model rationalizes for the first time the mechanisms that govern viscotaxis and its competition with durotaxis through a mathematical model.
Collapse
Affiliation(s)
- Pablo Saez
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, 08034 Barcelona, Spain; Institute of Mathematics of UPC-BarcelonaTech.-IMTech, Barcelona, Spain.
| | - Pallavi U Shirke
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), 400076 Mumbai, India
| | - Jyoti R Seth
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), 400076 Mumbai, India
| | | | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), 400076 Mumbai, India
| |
Collapse
|
30
|
Haidari R, Fowler WJ, Robinson SD, Johnson RT, Warren DT. Microvascular endothelial cells display organ-specific responses to extracellular matrix stiffness. Curr Res Physiol 2025; 8:100140. [PMID: 39967829 PMCID: PMC11833412 DOI: 10.1016/j.crphys.2025.100140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/21/2024] [Accepted: 01/24/2025] [Indexed: 02/20/2025] Open
Abstract
The extracellular matrix was originally thought of as simply a cellular scaffold but is now considered a key regulator of cell function and phenotype from which cells can derive biochemical and mechanical stimuli. Age-associated changes in matrix composition drive increases in matrix stiffness. Enhanced matrix stiffness promotes the progression of numerous diseases including cardiovascular disease, musculoskeletal disease, fibrosis, and cancer. Macrovascular endothelial cells undergo endothelial dysfunction in response to enhanced matrix stiffness. However, endothelial cells are highly heterogeneous, adopting structural and gene expression profiles specific to their organ of origin. Endothelial cells isolated from different vessels (i.e. arteries, veins or capillaries) respond differently to changes in substrate stiffness. It is unknown whether microvascular endothelial cells isolated from different organs also display organ-specific responses to substrate stiffness. In this study, we compare the response of microvascular endothelial cells isolated from both the mouse lung and mammary gland to a range of physiologically relevant substrate stiffnesses. We find that endothelial origin influences microvascular endothelial cell response to substrate stiffness in terms of both proliferation and migration speed. In lung-derived endothelial cells, proliferation is bimodal, where both physiologically soft and stiff substrates drive enhanced proliferation. Conversely, in mammary gland-derived endothelial cells, proliferation increases as substrate stiffness increases. Substrate stiffness also promotes enhanced endothelial migration. Enhanced stiffness drove greater increases in migration speed in mammary gland-derived than lung-derived endothelial cells. However, stiffness-induced changes in microvascular endothelial cell morphology were consistent between both cell lines, with substrate stiffness driving an increase in endothelial volume. Our research demonstrates the importance of considering endothelial origin in experimental design, especially when investigating how age-associated changes in matrix stiffness drive endothelial dysfunction and disease progression.
Collapse
Affiliation(s)
- Rana Haidari
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- School of Biology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Wesley J. Fowler
- Quadram Institute Bioscience, Norwich Research Park, NR4 7UQ, UK
| | - Stephen D. Robinson
- School of Biology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Quadram Institute Bioscience, Norwich Research Park, NR4 7UQ, UK
| | - Robert T. Johnson
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark
| | - Derek T. Warren
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
31
|
Cáceres-Calle D, Torre-Cea I, Marcos-Zazo L, Carrera-Aguado I, Guerra-Paes E, Berlana-Galán P, Muñoz-Félix JM, Sánchez-Juanes F. Integrins as Key Mediators of Metastasis. Int J Mol Sci 2025; 26:904. [PMID: 39940673 PMCID: PMC11816423 DOI: 10.3390/ijms26030904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Metastasis is currently becoming a major clinical concern, due to its potential to cause therapeutic resistance. Its development involves a series of phases that describe the metastatic cascade: preparation of the pre-metastatic niche, epithelial-mesenchymal transition, dissemination, latency and colonization of the new tissue. In the last few years, new therapeutic targets, such as integrins, are arising to face this disease. Integrins are transmembrane proteins found in every cell that have a key role in the metastatic cascade. They intervene in adhesion and intracellular signaling dependent on the extracellular matrix and cytokines found in the microenvironment. In this case, integrins can initiate the epithelial-mesenchymal transition, guide the formation of the pre-metastatic niche and increase tumor migration and survival. Integrins also take part in the tumor vascularization process necessary to sustain metastasis. This fact emphasizes the importance of inhibitory therapies capable of interfering with the function of integrins in metastasis.
Collapse
Affiliation(s)
- Daniel Cáceres-Calle
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Irene Torre-Cea
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Patricia Berlana-Galán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José M. Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
32
|
Jung S, Choi B, Lee M, Park S, Choi W, Yong H, Heo SE, Park Y, Lee JM, Lee ST, Hwang H, Kwon JS, Koh WG, Hong J. Bio-Orchestration of Cellular Organization and Human-Preferred Sensory Texture in Cultured Meat. ACS NANO 2025; 19:2809-2821. [PMID: 39772497 DOI: 10.1021/acsnano.4c15622] [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: 01/11/2025]
Abstract
For cultured meat to effectively replace traditional meat, it is essential to develop scaffolds that replicate key attributes of real meat, such as taste, nutrition, flavor, and texture. However, one of the significant challenges in replicating meat characteristics with scaffolds lies in the considerable gap between the stiffness preferred by cells and the textural properties desired by humans. To address this issue, we focused on the microscale environment conducive to cell growth and the macro-scale properties favored by humans. This led to the development of the adaptive bio-orchestrating anisotropic scaffold (ABS), which satisfies both cellular and human requirements. The ABS is produced using the anisotropic freeze-initiated ion coordination method, which sequentially aligns and enhances the fibril structure of food-derived proteins, effectively bridging the gap between cellular and culinary perspectives. Notably, the microenvironments of the ABS exhibited exceptional myoblast cell differentiation, with macro-scale 3D mechanical textures that are consistent regardless of the chewing direction, due to the aligned fibril and cell structure. The ABS containing bovine myotubes demonstrated a mechanical texture nearly identical to that of beef sirloins.
Collapse
Affiliation(s)
- Sungwon Jung
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Bumgyu Choi
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Milae Lee
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sohyeon Park
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Gordon Center for Medical Imaging, Boston, Massachusetts 02114, United States
| | - Woojin Choi
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyungseok Yong
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sung-Eun Heo
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Yeseul Park
- Digital OMICs Research Center, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Jeong Min Lee
- Department of Applied Animal Life Science, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341, Republic of Korea
| | - Seung Tae Lee
- Department of Applied Animal Life Science, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341, Republic of Korea
| | - Heeyoun Hwang
- Digital OMICs Research Center, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Bio-Analytical School, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jae-Sung Kwon
- Department and Research Institute of Dental Biomaterials and Bioengineering, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
33
|
Sheth M, Sharma M, Kongsomros S, Lehn M, Takebe T, Takiar V, Wise-Draper T, Chutipongtanate S, Esfandiari L. Matrix stiffness modulated release of spheroid-derived extracellular vesicles and discovery of Piezo1 cargo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632826. [PMID: 39868119 PMCID: PMC11760731 DOI: 10.1101/2025.01.13.632826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Augmented extracellular matrix (ECM) stiffness is a mechanical hallmark of cancer. Mechanotransduction studies have extensively probed the mechanisms by which ECM stiffness regulates intracellular communication. However, the influence of stiffness on intercellular communication aiding tumor progression in three-dimensional microenvironments remains unknown. Small extracellular vesicles (EVs) are communicators of altered biophysical cues to distant sites through EV-ECM interactions and EV-mediated recipient cell-ECM interactions. Here we demonstrate stiffness-mediated modulation of small EVs secretion and cargo from three-dimensional oral squamous cell carcinoma spheroids. Using a spheroid culture platform with varying matrix stiffness properties, we show that small EVs carry parental biomolecular cargo, including mechanosensitive Piezo1 ion channel and adhesion molecule CD44. We comprehensively validate the presence of both markers in our EV populations using proteomic and genetic analysis. Transcriptomic analysis of microRNA and long non-coding RNA cargo of small EVs released from soft and stiff ECM spheroids revealed enrichment of tumorigenic and metastatic profiles in EVs from stiff ECM cultures compared to that of soft ones. Gene set enrichment analysis of a comparative dataset obtained by overlaying spheroid mRNA and EV miRNA profiles identified key oncogenic pathways involved in cell-EV crosstalk in the spheroid model.
Collapse
|
34
|
Mitriashkin A, Yap JYY, Fernando EAK, Iyer NG, Grenci G, Fong ELS. Cell confinement by micropatterning induces phenotypic changes in cancer-associated fibroblasts. Acta Biomater 2025; 192:61-76. [PMID: 39637956 DOI: 10.1016/j.actbio.2024.12.007] [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: 08/02/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Recent advances in single-cell studies have revealed the vast transcriptomic heterogeneity of cancer-associated fibroblasts (CAFs), with each subset likely having unique roles in the tumor microenvironment. However, it is still unclear how different CAF subsets should be cultured in vitro to recapitulate their in vivo phenotype. The inherent plasticity of CAFs, or their ability to dynamically change their phenotype in response to different environmental stimuli, makes it highly challenging to induce and maintain a specific CAF state in vitro. In this study, we investigated how cell shape and confinement on two-dimensional culture substrates with different stiffnesses influence CAF transcriptomic profile and phenotype. Using micropatterning of polyacrylamide hydrogels to induce shape- and confinement-dependent changes in cell morphology, we observed that micropatterned CAFs exhibited phenotypic shifts towards more desmoplastic and inflammatory CAF subsets. Additionally, micropatterning enabled control over a range of CAF-specific markers and pathways. Lastly, we report how micropatterned and non-micropatterned CAFs respond differently to anti-cancer drugs, highlighting the importance of phenotype-oriented therapy that considers for CAF plasticity and regulatory networks. Control over CAF morphology offers a unique opportunity to establish highly robust CAF phenotypes in vitro, facilitating deeper understanding of CAF plasticity, heterogeneity, and development of novel therapeutic targets. STATEMENT OF SIGNIFICANCE: Cancer-associated fibroblasts (CAFs) are the dominant stromal cell type in many cancers, and recent studies have revealed that they are highly heterogeneous and comprise several subpopulations. It is still unclear how different subsets of CAFs should be cultured in vitro to recapitulate their in vivo phenotype. In this study, we investigated how cell shape and confinement affect CAF transcriptomic profile and phenotype. We report that micropatterned CAFs resemble desmoplastic and inflammatory CAF subsets observed in vivo and respond differently to anti-cancer drugs as compared to non-patterned CAFs. Control over CAF morphology enables the generation of highly robust CAF phenotypes in vitro, facilitating deeper understanding of CAF plasticity and heterogeneity.
Collapse
Affiliation(s)
- Aleksandr Mitriashkin
- Translational Tumor Engineering Laboratory, Department of Biomedical Engineering, National University of Singapore, Singapore 119276, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Josephine Yu Yan Yap
- Translational Tumor Engineering Laboratory, Department of Biomedical Engineering, National University of Singapore, Singapore 119276, Singapore
| | - Elekuttige Anton Kanishka Fernando
- Translational Tumor Engineering Laboratory, Department of Biomedical Engineering, National University of Singapore, Singapore 119276, Singapore
| | - N Gopalakrishna Iyer
- Cancer Therapeutics Research Laboratory, National Cancer Centre Singapore, Singapore 168583, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
| | - Gianluca Grenci
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Eliza Li Shan Fong
- Translational Tumor Engineering Laboratory, Department of Biomedical Engineering, National University of Singapore, Singapore 119276, Singapore; Cancer Science Institute, National University of Singapore, Singapore 117599, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore 117456, Singapore.
| |
Collapse
|
35
|
Narain R, Muncie-Vasic JM, Weaver VM. Forcing the code: tension modulates signaling to drive morphogenesis and malignancy. Genes Dev 2025; 39:163-181. [PMID: 39638568 PMCID: PMC11789492 DOI: 10.1101/gad.352110.124] [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: 12/07/2024]
Abstract
Development and disease are regulated by the interplay between genetics and the signaling pathways stimulated by morphogens, growth factors, and cytokines. Experimental data highlight the importance of mechanical force in regulating embryonic development, tissue morphogenesis, and malignancy. Force not only sculpts tissue movements to drive embryogenesis and morphogenesis but also modifies the context of biochemical signaling and gene expression to regulate cell and tissue fate. Not surprisingly, experiments have demonstrated that perturbations in cell tension drive malignancy and metastasis by altering biochemical signaling and gene expression through modifications in cytoskeletal tension, transmembrane receptor structure and function, and organelle phenotype that enhance cell growth and survival, alter metabolism, and foster cell migration and invasion. At the tissue level, tumor-associated forces disrupt cell-cell adhesions to perturb tissue organization, compromise vascular integrity to induce hypoxia, and interfere with antitumor immunity to foster metastasis and treatment resistance. Exciting new approaches now exist with which to clarify the relationship between mechanotransduction, biochemical signaling, and gene expression in development and disease. Indeed, gaining insight into these interactions is essential to unravel molecular mechanisms that regulate development and clarify the molecular basis of cancer.
Collapse
Affiliation(s)
- Radhika Narain
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California 94720, USA
| | | | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA;
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California 94143
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
36
|
Lan H, Tan XHM, Le MTT, Chien HY, Zheng R, Rowat AC, Teitell MA, Chiou PY. Optomagnetic Micromirror Arrays for Mapping Large Area Stiffness Distributions of Biomimetic Materials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406389. [PMID: 39614709 PMCID: PMC11710979 DOI: 10.1002/smll.202406389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/17/2024] [Indexed: 12/01/2024]
Abstract
A new device termed "Optomagnetic Micromirror Arrays" (OMA) is demonstrated capable of mapping the stiffness distribution of biomimetic materials across a 5.1 mm × 7.2 mm field of view with cellular resolution. The OMA device comprises an array of 50 000 magnetic micromirrors with optical grating structures embedded beneath an elastic PDMS film, with biomimetic materials affixed on top. Illumination of a broadband white light beam onto these micromirrors results in the reflection of microscale rainbow light rays on each micromirror. When a magnetic field is applied, it causes each micromirror to tilt differently depending on the local stiffness of the biomimetic materials. Through imaging these micromirrors with low N.A. optics, a specific narrow band of reflection light rays from each micromirror is captured. Changing a micromirror's tilt angle also alters the color spectrum it reflects back to the imaging system and the color of the micromirror image it represents. As a result, OMA can infer the local stiffness of the biomimetic materials through the color change detected on each micromirror. OMA offers the potential for high-throughput stiffness mapping at the tissue-level while maintaining spatial resolution at the cellular level.
Collapse
Affiliation(s)
- Hsin Lan
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xing Haw Marvin Tan
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, 138632, Republic of Singapore
| | - Minh-Tam Tran Le
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Hao-Yu Chien
- Department of Electrical and Computer Enigeering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ruoda Zheng
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Amy C Rowat
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology & Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael A Teitell
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Pei-Yu Chiou
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
37
|
Steinbeck L, Paul R, Litke J, Karkoszka I, Wiese GP, Linkhorst J, De Laporte L, Wessling M. Hierarchically Structured and Tunable Hydrogel Patches: Design, Characterization, and Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407311. [PMID: 39568251 PMCID: PMC11753498 DOI: 10.1002/smll.202407311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/25/2024] [Indexed: 11/22/2024]
Abstract
Recent studies show the importance of hydrogel geometry for various applications, such as encoding, micromachines, or tissue engineering. However, fabricating hydrogel structures with micrometer-sized features, advanced geometry, and precise control of porosity remains challenging. This work presents hierarchically structured hydrogels, so-called hydrogel patches, with internally deviating regions on a micron-scale. These regions are defined in a one-step, high-throughput fabrication process via stop-flow lithography. Between the specified projection pattern during fabrication, an interconnecting lower crosslinked and more porous hydrogel network forms, resulting in at least two degrees of crosslinking within the patches. A detailed investigation of patch formation is performed for two material systems and pattern variations, revealing basic principles for reliable patch formation. In addition to the two defined crosslinked regions, further regions are implemented in the patches by adapting the pattern accordingly. The variations in pattern geometry impact the mechanical characteristics of the hydrogel patches, which display pattern-dependent compression behavior due to predefined compression points. Cell culture on patches, as one possible application, reveals that the patch pattern determines the cell area of L929 mouse fibroblasts. These results introduce hierarchically structured hydrogel patches as a promising and versatile platform system with high customizability.
Collapse
Affiliation(s)
- Lea Steinbeck
- Chemical Process Engineering AVT.CVTRWTH Aachen UniversityForckenbeckstraße 5152074AachenGermany
| | - Richard Paul
- Chemical Process Engineering AVT.CVTRWTH Aachen UniversityForckenbeckstraße 5152074AachenGermany
- DWI – Leibniz‐Institute for Interactive Materials e. V.Forckenbeckstraße 5052074AachenGermany
| | - Julia Litke
- Chemical Process Engineering AVT.CVTRWTH Aachen UniversityForckenbeckstraße 5152074AachenGermany
| | - Isabel Karkoszka
- Chemical Process Engineering AVT.CVTRWTH Aachen UniversityForckenbeckstraße 5152074AachenGermany
| | - G. Philip Wiese
- Chemical Process Engineering AVT.CVTRWTH Aachen UniversityForckenbeckstraße 5152074AachenGermany
| | - John Linkhorst
- Chemical Process Engineering AVT.CVTRWTH Aachen UniversityForckenbeckstraße 5152074AachenGermany
- Present address:
Process Engineering of Electrochemical SystemsTechnical University of DarmstadtOtto‐Berndt‐Str. 264287DarmstadtGermany
| | - Laura De Laporte
- DWI – Leibniz‐Institute for Interactive Materials e. V.Forckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 1‐252074AachenGermany
- Center for Biohybrid Medical Systems (CBMS)Advanced Materials for Biomedicine (AMB)Institute of Applied Medical Engineering (AME)University Hospital RWTH AachenForckenbeckstraße 5552074AachenGermany
| | - Matthias Wessling
- Chemical Process Engineering AVT.CVTRWTH Aachen UniversityForckenbeckstraße 5152074AachenGermany
- DWI – Leibniz‐Institute for Interactive Materials e. V.Forckenbeckstraße 5052074AachenGermany
| |
Collapse
|
38
|
Cassani M, Fernandes S, Pagliari S, Cavalieri F, Caruso F, Forte G. Unraveling the Role of the Tumor Extracellular Matrix to Inform Nanoparticle Design for Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409898. [PMID: 39629891 PMCID: PMC11727388 DOI: 10.1002/advs.202409898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Indexed: 01/14/2025]
Abstract
The extracellular matrix (ECM)-and its mechanobiology-regulates key cellular functions that drive tumor growth and development. Accordingly, mechanotherapy is emerging as an effective approach to treat fibrotic diseases such as cancer. Through restoring the ECM to healthy-like conditions, this treatment aims to improve tissue perfusion, facilitating the delivery of chemotherapies. In particular, the manipulation of ECM is gaining interest as a valuable strategy for developing innovative treatments based on nanoparticles (NPs). However, further progress is required; for instance, it is known that the presence of a dense ECM, which hampers the penetration of NPs, primarily impacts the efficacy of nanomedicines. Furthermore, most 2D in vitro studies fail to recapitulate the physiological deposition of matrix components. To address these issues, a comprehensive understanding of the interactions between the ECM and NPs is needed. This review focuses on the main features of the ECM and its complex interplay with NPs. Recent advances in mechanotherapy are discussed and insights are offered into how its combination with nanomedicine can help improve nanomaterials design and advance their clinical translation.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Stefania Pagliari
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
39
|
Xue SL. Tissue stresses caused by invasive tumour: a biomechanical model. J R Soc Interface 2025; 22:20240797. [PMID: 39837483 PMCID: PMC11750364 DOI: 10.1098/rsif.2024.0797] [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: 07/06/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Malignant tumorigenesis is a complex process involving growth, invasion and mechanical deformation of a cancerous tissue. In this paper, a biomechanical model is proposed to couple the mechanical and biological mechanisms governing invasive tumour development. As an example, this model is applied to investigate the spatio-temporal evolution of tissue stresses in an invasive tumour spheroid and its host tissue. I show that cancer invasiveness lowers the compressive tissue stresses and blurs the stress distribution across the cancerous-normal tissue boundary, both consistent with experimental observations. Importantly, with the steady propagation of the cancerous region driven by persistent cancer invasion, tumour stresses are predicted to saturate rather than keep increasing as in benign tumour growth. The model is further used to analyse the deformation and stress state of a cancerous tissue being cut into two pieces, and reproduces the bulge of the cut surface observed in experiments. I hope this study can pave the way for the quantitative evaluation of mechanical states in cancer.
Collapse
Affiliation(s)
- Shi-Lei Xue
- Department of Materials Science and Engineering, School of Engineering, Westlake University, Hangzhou, Zhejiang310030, People’s Republic of China
| |
Collapse
|
40
|
Rauer SB, Stüwe L, Steinbeck L, de Toledo MAS, Fischer G, Wennemaring S, Marschick J, Koschmieder S, Wessling M, Linkhorst J. Cell Adhesion and Local Cytokine Control on Protein-Functionalized PNIPAM-co-AAc Hydrogel Microcarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2404183. [PMID: 39535368 PMCID: PMC11735893 DOI: 10.1002/smll.202404183] [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: 05/23/2024] [Revised: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Achieving adequate cell densities remains a major challenge in establishing economic biotechnological and biomedical processes. A possible remedy is microcarrier-based cultivation in stirred-tank bioreactors (STBR), which offers a high surface-to-volume ratio, appropriate process control, and scalability. However, despite their potential, commercial microcarriers are currently limited to material systems featuring unnatural mechanical properties and low adaptability. Because matrix stiffness and ligand presentation impact phenotypical attributes, differentiation potential, and genetic stability, biotechnological processes can significantly benefit from microcarrier systems tailorable toward cell-type specific requirements. This study introduces hydrogel particles co-polymerized from poly(N-isopropylacrylamide) (PNIPAM) and acrylic acid (AAc) as a platform technology for cell expansion. The resulting microcarriers exhibit an adjustable extracellular matrix-like softness, an adaptable gel charge, and functional carboxyl groups, allowing electrostatic and covalent coupling of cell adhesive and cell fate-modulating proteins. These features enable the attachment and growth of L929 mouse fibroblast cells in static microtiter plates and dynamic STBR cultivations while also providing vital growth factors, such as interleukin-3, to myeloblast-like 32D cells over 20 days of cultivation. The study explores the effects of different educt compositions on cell-particle interactions and reveals that PNIPAM-co-AAc microcarriers can provide both covalently coupled and diffusively released cytokine to adjacent cells.
Collapse
Affiliation(s)
- Sebastian Bernhard Rauer
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- DWI ‐ Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - Lucas Stüwe
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Lea Steinbeck
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Marcelo Augusto Szymanski de Toledo
- Department of HematologyOncology, Hemostaseology, and Stem Cell TransplantationFaculty of MedicineRWTH Aachen University52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD)52074AachenGermany
| | - Gereon Fischer
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Simon Wennemaring
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Jonas Marschick
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Steffen Koschmieder
- Department of HematologyOncology, Hemostaseology, and Stem Cell TransplantationFaculty of MedicineRWTH Aachen University52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD)52074AachenGermany
| | - Matthias Wessling
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- DWI ‐ Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - John Linkhorst
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- Process Engineering of Electrochemical SystemsDepartment of Mechanical EngineeringTechnical University of DarmstadtOtto‐Berndt‐Str. 264287DarmstadtGermany
| |
Collapse
|
41
|
Pal K. Unravelling molecular mechanobiology using DNA-based fluorogenic tension sensors. J Mater Chem B 2024; 13:37-53. [PMID: 39564891 DOI: 10.1039/d4tb01858c] [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: 11/21/2024]
Abstract
Investigations of the biological system have revealed many principles that govern regular life processes. Recently, the analysis of tiny mechanical forces associated with many biological processes revealed their significance in understanding biological functions. Consequently, this piqued the interest of researchers, and a series of technologies have been developed to understand biomechanical cues at the molecular level. Notable techniques include single-molecule force spectroscopy, traction force microscopy, and molecular tension sensors. Well-defined double-stranded DNA structures could possess programmable mechanical characteristics, and hence, they have become one of the central molecules in molecular tension sensor technology. With the advancement of DNA technology, DNA or nucleic acid-based robust tension sensors offer the possibility of understanding mechanobiology in the bulk to single-molecule level range with desired spatiotemporal resolution. This review presents a comprehensive account of molecular tension sensors with a special emphasis on DNA-based fluorogenic tension sensors. Along with a detailed discussion on irreversible and reversible DNA-based tension sensors and their application in super-resolution microscopy, a discussion on biomolecules associated with cellular mechanotransduction and key findings in the field are included. This review ends with an elaborate discussion on the current challenges and future prospects of molecular tension sensors.
Collapse
Affiliation(s)
- Kaushik Pal
- Biophysical Chemistry Laboratory, Department of Chemistry, Indian Institute of Technology Tirupati, Yerpedu, Tirupati, AP-517619, India.
| |
Collapse
|
42
|
Paradiso A, Volpi M, Martinez DC, Jaroszewicz J, Costantini M, Swieszkowski W. Engineering Biomimetic Microvascular Capillary Networks in Hydrogel Fibrous Scaffolds via Microfluidics-Assisted Co-Axial Wet-Spinning. ACS APPLIED MATERIALS & INTERFACES 2024; 16:65927-65941. [PMID: 39566902 PMCID: PMC11622188 DOI: 10.1021/acsami.4c15221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
The microvascular bed plays a crucial role in establishing nutrient exchange and waste removal, as well as maintaining tissue metabolic activity in the human body. However, achieving microvascularization of engineered 3D tissue constructs is still an unsolved challenge. In this work, we developed biomimetic cell-laden hydrogel microfibers recapitulating oriented microvascular capillary-like networks by using a 3D bioprinting technique combined with microfluidics-assisted coaxial wet-spinning. Highly packed and aligned bundles embedding a coculture of human bone marrow-derived mesenchymal stem cells (MSCs) and human umbilical vein endothelial cells (HUVECs) were produced by simultaneously extruding two different bioinks. To this aim, core-shell fibers were wet-spun in a coagulation bath to collect the scaffolds later on a rotary drum. Initially, the versatility of the proposed system was assessed for the extrusion of multimaterial core-shell hydrogel fibers. Subsequently, the platform was validated for the in vitro biofabrication of samples promoting optimal cell alignment along the fiber axis. After 3 weeks of culture, such fiber configuration resulted in the development of an oriented capillary-like network within the fibrin-based core and in the endothelial-specific CD31 marker expression upon MSC/HUVEC maturation. Synergistically, the vertical arrangement of the coaxial nozzle coupled with the rotation of the fiber collector facilitated the rapid creation of tightly packed bundles characterized by a dense, oriented, and extensively branched capillary network. Notably, such findings suggest that the proposed biofabrication strategy can be used for the microvascularization of tissue-specific 3D constructs.
Collapse
Affiliation(s)
- Alessia Paradiso
- Faculty of Materials Sciences and Engineering, Warsaw University of Technology, Warsaw 02-507, Poland
| | - Marina Volpi
- Faculty of Materials Sciences and Engineering, Warsaw University of Technology, Warsaw 02-507, Poland
| | - Diana C Martinez
- Faculty of Materials Sciences and Engineering, Warsaw University of Technology, Warsaw 02-507, Poland
| | - Jakub Jaroszewicz
- Faculty of Materials Sciences and Engineering, Warsaw University of Technology, Warsaw 02-507, Poland
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw 01-224, Poland
| | - Wojciech Swieszkowski
- Faculty of Materials Sciences and Engineering, Warsaw University of Technology, Warsaw 02-507, Poland
| |
Collapse
|
43
|
Hollander S, Guo Y, Wolfenson H, Zaritsky A. Spatiotemporal analysis of F-actin polymerization with micropillar arrays reveals synchronization between adhesion sites. Mol Biol Cell 2024; 35:br23. [PMID: 39441710 PMCID: PMC11656478 DOI: 10.1091/mbc.e24-06-0276] [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: 06/25/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
We repurposed micropillar arrays to quantify spatiotemporal inter-adhesion communication. Following the observation that integrin adhesions formed around pillar tops we relied on the precise repetitive spatial control of the pillars to reliably monitor F-actin dynamics in mouse embryonic fibroblasts as a model for spatiotemporal adhesion-related intracellular signaling. Using correlation-based analyses, we revealed localized information flows propagating between adjacent pillars that were integrated over space and time to synchronize the adhesion dynamics within the entire cell. Probing the mechanical regulation, we discovered that stiffer pillars or partial actomyosin contractility inhibition enhances inter-adhesion F-actin synchronization, and that inhibition of Arp2/3, but not formin, reduces synchronization. Our results suggest that adhesions can communicate and highlight the potential of using micropillar arrays as a tool to measure spatiotemporal intracellular signaling.
Collapse
Affiliation(s)
- Sarit Hollander
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yuanning Guo
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Assaf Zaritsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
44
|
Zanotelli MR, Miller JP, Wang W, Ortiz I, Tahon E, Bordeleau F, Reinhart-King CA. Tension directs cancer cell migration over fiber alignment through energy minimization. Biomaterials 2024; 311:122682. [PMID: 38959532 DOI: 10.1016/j.biomaterials.2024.122682] [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: 11/07/2023] [Revised: 06/06/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
Cell migration during many fundamental biological processes including metastasis requires cells to traverse tissue with heterogeneous mechanical cues that direct migration as well as determine force and energy requirements for motility. However, the influence of discrete structural and mechanical cues on migration remains challenging to determine as they are often coupled. Here, we decouple the pro-invasive cues of collagen fiber alignment and tension to study their individual impact on migration. When presented with both cues, cells preferentially travel in the axis of tension against fiber alignment. Computational and experimental data show applying tension perpendicular to alignment increases potential energy stored within collagen fibers, lowering requirements for cell-induced matrix deformation and energy usage during migration compared to motility in the direction of fiber alignment. Energy minimization directs migration trajectory, and tension can facilitate migration against fiber alignment. These findings provide a conceptual understanding of bioenergetics during migration through a fibrous matrix.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joseph P Miller
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Ismael Ortiz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Elise Tahon
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, G1R 3S3, Canada
| | - Francois Bordeleau
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, G1R 3S3, Canada; Département de Biologie Moléculaire, de Biochimie Médicale et de Pathologie, Université Laval, Québec, Canada, G1V 0A6.
| | | |
Collapse
|
45
|
Ferrara B, Bourgoin-Voillard S, Habert D, Vallée B, Nicolas-Boluda A, Simanic I, Seve M, Vingert B, Gazeau F, Castellano F, Cohen J, Courty J, Cascone I. Matrix stiffness regulates the protein profile of extracellular vesicles of pancreatic cancer cell lines. Proteomics 2024; 24:e2400058. [PMID: 39279557 DOI: 10.1002/pmic.202400058] [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: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024]
Abstract
The fibrotic stroma characterizing pancreatic ductal adenocarcinoma (PDAC) derives from a progressive tissue rigidification, which induces epithelial mesenchymal transition and metastatic dissemination. The aim of this study was to investigate the influence of matrix stiffness on PDAC progression by analyzing the proteome of PDAC-derived extracellular vesicles (EVs). PDAC cell lines (mPDAC and KPC) were grown on synthetic supports with a stiffness close to non-tumor (NT) or tumor tissue (T), and the protein expression levels in cell-derived EVs were analyzed by a quantitative MSE label-free mass spectrometry approach. Our analysis figured out 15 differentially expressed proteins (DEPs) in mPDAC-EVs and 20 DEPs in KPC-EVs in response to matrix rigidification. Up-regulated proteins participate to the processes of metabolism, matrix remodeling, and immune response, altogether hallmarks of PDAC progression. A multimodal network analysis revealed that the majority of DEPs are strongly related to pancreatic cancer. Interestingly, among DEPs, 11 related genes (ACTB/ANXA7/C3/IGSF8/LAMC1/LGALS3/PCD6IP/SFN/TPM3/VARS/YWHAZ) for mPDAC-EVs and 9 (ACTB/ALDH2/GAPDH/HNRNPA2B/ITGA2/NEXN/PKM/RPN1/S100A6) for KPC-EVs were significantly overexpressed in tumor tissues according to gene expression profiling interaction analysis (GEPIA). Concerning the potential clinical relevance of these data, the cluster of ACTB, ITGA2, GAPDH and PKM genes displayed an adverse effect (p < 0.05) on the overall survival of PDAC patients.
Collapse
Affiliation(s)
- Benedetta Ferrara
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sandrine Bourgoin-Voillard
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, CHU Grenoble Alpes, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, LBFA et BEeSy, Inserm, U1055, CHU Grenoble Alpes, PROMETHEE Proteomic Platform, Grenoble, France
| | - Damien Habert
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - Benoit Vallée
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - Alba Nicolas-Boluda
- Matière et Systèmes Complexes MSC, CNRS, Université Paris Cité, Paris, France
| | - Isidora Simanic
- Modèles de cellules souches malignes et therapeutiques, INSERM UMR-S 935, Université Paris-Saclay, Villejuif, France
| | - Michel Seve
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, CHU Grenoble Alpes, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, LBFA et BEeSy, Inserm, U1055, CHU Grenoble Alpes, PROMETHEE Proteomic Platform, Grenoble, France
| | - Benoit Vingert
- Etablissement Français du Sang, Créteil, France
- Inserm, U955, Equipe 2, Créteil, France
| | - Florence Gazeau
- Matière et Systèmes Complexes MSC, CNRS, Université Paris Cité, Paris, France
| | - Flavia Castellano
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - José Cohen
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Centre d'investigation clinique Biotherapie, Créteil, France
| | - José Courty
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Centre d'investigation clinique Biotherapie, Créteil, France
| | - Ilaria Cascone
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Centre d'investigation clinique Biotherapie, Créteil, France
| |
Collapse
|
46
|
Barrett L, Coopman K. Cell microencapsulation techniques for cancer modelling and drug discovery. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:345-354. [PMID: 38829715 DOI: 10.1080/21691401.2024.2359996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
Cell encapsulation into spherical microparticles is a promising bioengineering tool in many fields, including 3D cancer modelling and pre-clinical drug discovery. Cancer microencapsulation models can more accurately reflect the complex solid tumour microenvironment than 2D cell culture and therefore would improve drug discovery efforts. However, these microcapsules, typically in the range of 1 - 5000 µm in diameter, must be carefully designed and amenable to high-throughput production. This review therefore aims to outline important considerations in the design of cancer cell microencapsulation models for drug discovery applications and examine current techniques to produce these. Extrusion (dripping) droplet generation and emulsion-based techniques are highlighted and their suitability to high-throughput drug screening in terms of tumour physiology and ease of scale up is evaluated.
Collapse
Affiliation(s)
- Lisa Barrett
- Department of Chemical Engineering, School of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, UK
| | - Karen Coopman
- Department of Chemical Engineering, School of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, UK
| |
Collapse
|
47
|
Avery K, Chen X. Integration of bioprinting advances and biomechanical strategies for in vitrolung modelling. Biofabrication 2024; 17:012006. [PMID: 39536463 DOI: 10.1088/1758-5090/ad91e2] [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: 06/20/2024] [Accepted: 11/13/2024] [Indexed: 11/16/2024]
Abstract
The recent occurrence of the Covid-19 pandemic and frequent wildfires have worsened pulmonary diseases and raised the urgent need for investigating host-pathogen interactions and advancing drug and vaccine therapies. Historically, research and experimental studies have relied on two-dimensional cell culture dishes and/or animal models, which suffer from physiological differences from the human lung. More recently, there has been investigation into the use of lung-on-a-chip models and organoids, while the use of bioprinting technologies has also emerged to fabricate three-dimensional constructs or lung models with enhanced physiological relevance. Concurrently, achievements have also been made to develop biomimetic strategies for simulating thein vivobiomechanical conditions induced by lung breathing, though challenges remain with incorporating these strategies with bioprinted models. Bioprinted models combined with advanced biomimetic strategies would represent a promising approach to advance disease discovery and therapeutic development. As inspired, this article briefly reviews the recent progress of both bioprintedin vitrolung models and biomechanical strategies, with a focus on native lung tissue microstructure and biomechanical properties, bioprinted constructs, and biomimetic strategies to mimic the native environment. This article also urges that the integration of bioprinting advances and biomimetic strategies would be essential to achieve synergistic effects forin vitrolung modelling. Key issues and challenges are also identified and discussed along with recommendations for future research.
Collapse
Affiliation(s)
- Kathryn Avery
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
48
|
Wang TC, Abolghasemzade S, McKee BP, Singh I, Pendyala K, Mohajeri M, Patel H, Shaji A, Kersey AL, Harsh K, Kaur S, Dollahon CR, Chukkapalli S, Lele PP, Conway DE, Gaharwar AK, Dickinson RB, Lele TP. Matrix stiffness drives drop like nuclear deformation and lamin A/C tension-dependent YAP nuclear localization. Nat Commun 2024; 15:10151. [PMID: 39578439 PMCID: PMC11584751 DOI: 10.1038/s41467-024-54577-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Extracellular matrix (ECM) stiffness influences cancer cell fate by altering gene expression. Previous studies suggest that stiffness-induced nuclear deformation may regulate gene expression through YAP nuclear localization. We investigated the role of the nuclear lamina in this process. We show that the nuclear lamina exhibits mechanical threshold behavior: once unwrinkled, the nuclear lamina is inextensible. A computational model predicts that the unwrinkled lamina is under tension, which is confirmed using a lamin tension sensor. Laminar unwrinkling is caused by nuclear flattening during cell spreading on stiff ECM. Knockdown of lamin A/C eliminates nuclear surface tension and decreases nuclear YAP localization. These findings show that nuclear deformation in cells conforms to the nuclear drop model and reveal a role for lamin A/C tension in controlling YAP localization in cancer cells.
Collapse
Affiliation(s)
- Ting-Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Samere Abolghasemzade
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Brendan P McKee
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Ishita Singh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kavya Pendyala
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Hailee Patel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Aakansha Shaji
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Anna L Kersey
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kajol Harsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Simran Kaur
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Christina R Dollahon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Sasanka Chukkapalli
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Pushkar P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Tanmay P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
49
|
Li T, Cheburkanov V, Yakovlev VV, Agarwal GS, Scully MO. Harnessing quantum light for microscopic biomechanical imaging of cells and tissues. Proc Natl Acad Sci U S A 2024; 121:e2413938121. [PMID: 39480851 PMCID: PMC11551316 DOI: 10.1073/pnas.2413938121] [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: 07/11/2024] [Accepted: 09/24/2024] [Indexed: 11/02/2024] Open
Abstract
The biomechanical properties of cells and tissues play an important role in our fundamental understanding of the structures and functions of biological systems at both the cellular and subcellular levels. Recently, Brillouin microscopy, which offers a label-free spectroscopic means of assessing viscoelastic properties in vivo, has emerged as a powerful way to interrogate those properties on a microscopic level in living tissues. However, susceptibility to photodamage and photobleaching, particularly when high-intensity laser beams are used to induce Brillouin scattering, poses a significant challenge. This article introduces a transformative approach designed to mitigate photodamage in biological and biomedical studies, enabling nondestructive, label-free assessments of mechanical properties in live biological samples. By leveraging quantum-light-enhanced stimulated Brillouin scattering (SBS) imaging contrast, the signal-to-noise ratio is significantly elevated, thereby increasing sample viability and extending interrogation times without compromising the integrity of living samples. The tangible impact of this methodology is evidenced by a notable three-fold increase in sample viability observed after subjecting the samples to three hours of continuous squeezed-light illumination, surpassing the traditional coherent light-based approaches. The quantum-enhanced SBS imaging holds promise across diverse fields, such as cancer biology and neuroscience where preserving sample vitality is of paramount significance. By mitigating concerns regarding photodamage and photobleaching associated with high-intensity lasers, this technological breakthrough expands our horizons for exploring the mechanical properties of live biological systems, paving the way for an era of research and clinical applications.
Collapse
Affiliation(s)
- Tian Li
- Department of Chemistry and Physics, The University of Tennessee, Chattanooga, TN37403
- The University of Tennessee Research Institute, The University of Tennessee, Chattanooga, TN37403
| | - Vsevolod Cheburkanov
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
| | - Vladislav V. Yakovlev
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
- Institute for Quantum Science and Engineering, Department of Physics and Astronomy, Texas A&M University, College Station, TX77843
| | - Girish S. Agarwal
- Institute for Quantum Science and Engineering, Department of Physics and Astronomy, Texas A&M University, College Station, TX77843
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX77843
| | - Marlan O. Scully
- Institute for Quantum Science and Engineering, Department of Physics and Astronomy, Texas A&M University, College Station, TX77843
| |
Collapse
|
50
|
Wu X, Kong Y, Yi Y, Xu S, Chen J, Chen J, Jin P. Label-Free Monitoring of Endometrial Cancer Progression Using Multiphoton Microscopy. Ann Biomed Eng 2024; 52:3113-3124. [PMID: 38960975 PMCID: PMC11511711 DOI: 10.1007/s10439-024-03574-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Endometrial cancer is the most common gynecological cancer in the developed world. However, the accuracy of current diagnostic methods is still unsatisfactory and time-consuming. Here, we presented an alternate approach to monitoring the progression of endometrial cancer via multiphoton microscopy imaging and analysis of collagen, which is often overlooked in current endometrial cancer diagnosis protocols but can offer a crucial signature in cancer biology. Multiphoton microscopy (MPM) based on the second-harmonic generation and two-photon excited fluorescence was introduced to visualize the microenvironment of endometrium in normal, hyperplasia without atypia, atypical hyperplasia, and endometrial cancer specimens. Furthermore, automatic image analysis based on the MPM image processing algorithm was used to quantify the differences in the collagen morphological features among them. MPM enables the visualization of the morphological details and alterations of the glands in the development process of endometrial cancer, including irregular changes in the structure of the gland, increased ratio of the gland to the interstitium, and atypical changes in the glandular epithelial cells. Moreover, the destructed basement membrane caused by gland proliferation and fusion is clearly shown in SHG images, which is a key feature for identifying endometrial cancer progression. Quantitative analysis reveals that the formation of endometrial cancer is accompanied by an increase in collagen fiber length and width, a progressive linearization and loosening of interstitial collagen, and a more random arrangement of interstitial collagen. Observation and quantitative analysis of interstitial collagen provide invaluable information in monitoring the progression of endometrial cancer. Label-free multiphoton imaging reported here has the potential to become an in situ histological tool for effective and accurate early diagnosis and detection of malignant lesions in endometrial cancer.
Collapse
Affiliation(s)
- Xuzhen Wu
- Department of Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Shandong University, Shenzhen, 518028, China
| | - Yanqing Kong
- Department of Pathology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, 518028, China
| | - Yu Yi
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, 350117, China
| | - Shuoyu Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jianhua Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, 350117, China.
- College of Life Science, Fujian Normal University, Fuzhou, 350117, China.
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, 350117, China.
| | - Ping Jin
- Department of Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Shandong University, Shenzhen, 518028, China.
| |
Collapse
|