1
|
Deng H, Xiao Q, Xu X, Zhang L, Zhang Y. Quercetin Inhibits Gastric Cancer Progression via FAM198B/MAPK Pathway Modulation. Pharmgenomics Pers Med 2025; 18:115-141. [PMID: 40390771 PMCID: PMC12087595 DOI: 10.2147/pgpm.s511324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Background The family with the sequence similarity 198 member B (FAM198B) has been found to contribute to the progression of gastric cancer (GC). However, the role and molecular mechanism of FAM198B in GC remains poorly understood. This work found a link between FAM198B and quercetin, and the regulatory effect of FAM198B on the MAPK pathway of GC. Methods FAM198B expression was identified through multiple public data sets and verified in clinical tissue samples. The associations between FAM198B and the prognosis of patients with GC were analyzed via the Kaplan‒Meier plotter and Cox regression analysis. Gene set enrichment analysis, coexpressed genes, and RNA sequencing were used to explore the related functions and signaling pathways of FAM198B in GC. In vitro assays assessed the effects of FAM198B knockdown on GC cells. FAM198B was found as a quercetin target by the HERB database and in vitro assays. Results FAM198B was highly expressed in tissues from GC patients (p<0.001) and was positively associated with poor prognosis (p<0.001) and immune cell infiltration in GC patients. FAM198B knockdown inhibited the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of GC cells (all p<0.05). In addition, FAM198B knockdown decreased the phosphorylation of p-Erk1/2 and p-p38 in GC cells (all p<0.01). Quercetin inhibited FAM198B expression and the phosphorylation of p-Erk1/2 and p-p38 in GC cells (all p<0.05). Conclusion Quercetin inhibits the proliferation, migration, invasion, and EMT of GC cells by inhibiting the FAM198B/MAPK signaling pathway. These discoveries lay the groundwork for developing the treatment of GC by quercetin and targeting FAM198B. In the future, more preclinical and clinical studies are needed to confirm the efficacy and safety of quercetin and target FAM198B in GC.
Collapse
Affiliation(s)
- Hongyang Deng
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Qi Xiao
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xiaodong Xu
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Lingyi Zhang
- Department of Liver Disease, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Youcheng Zhang
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
2
|
Liang Z, Ye Y, Deng Z, Lan H, Liu C, Xu Y, Fan M, Liu Z, Wu P, An L, Wang C. CHPF2 as a novel biomarker and ponicidin as a potential therapeutic agent in hepatocellular carcinoma. Pharmacol Res 2025; 215:107698. [PMID: 40107635 DOI: 10.1016/j.phrs.2025.107698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Hepatocellular carcinoma (HCC) was associated with high morbidity and mortality, representing a significant health challenge. Chondroitin sulfate (CS), a glycosaminoglycan composed of glucuronic acid and N-acetylgalactosamine, is implicated in HCC progression through its role in cancer cell migration and proliferation as well as interactions with cell surface receptors integrin β-1 and CD44. Chondroitin polymerization factor 2 (CHPF2), the key to CS synthesis, has an undefined role in HCC. Our study aims to demonstrate that decreasing CHPF2 enzyme activity can inhibit the migration and proliferation of HCC cells. Bioinformatics analysis and in vitro experiments on clinical HCC samples confirmed the knockdown of CHPF2 inhibited HCC cell proliferation and migration. We further explored Rabdosia rubescens, a plant used in cancer therapy, for its potential to modulate CHPF2. Structural biology and ligand fishing identified ponicidin, a compound that significantly suppresses HCC cell growth and migration in both in vitro and in vivo models. These findings propose CHPF2 as a novel biomarker and ponicidin as a potential therapeutic agent for HCC management.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- Cell Proliferation/drug effects
- Cell Movement/drug effects
- Animals
- Cell Line, Tumor
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Mice, Nude
- Male
- MARVEL Domain-Containing Proteins/metabolism
- MARVEL Domain-Containing Proteins/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Mice, Inbred BALB C
- Hep G2 Cells
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
Collapse
Affiliation(s)
- Zuhui Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Yingyi Ye
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Zhihong Deng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Huan Lan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Caihong Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Yuanhang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Minqi Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Peng Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| | - Lin An
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| |
Collapse
|
3
|
Martinez-Marin D, Stroman GC, Fulton CJ, Pruitt K. Frizzled receptors: gatekeepers of Wnt signaling in development and disease. Front Cell Dev Biol 2025; 13:1599355. [PMID: 40376615 PMCID: PMC12078226 DOI: 10.3389/fcell.2025.1599355] [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: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Frizzled (FZD) receptors are a subset of G-protein-coupled receptors (GPCRs), the largest class of human cell surface receptors and a major target of FDA-approved drugs. Activated by Wnt ligands, FZDs regulate key cellular processes such as proliferation, differentiation, and polarity, positioning them at the intersection of developmental biology and disease, including cancer. Despite their significance, FZD signaling remains incompletely understood, particularly in distinguishing receptor-specific roles across canonical and non-canonical Wnt pathways. Challenges include defining ligand-receptor specificity, elucidating signal transduction mechanisms, and understanding the influence of post translational modifications and the cellular context. Structural dynamics, receptor trafficking, and non-canonical signaling contributions also remain areas of active investigation. Recent advances in structural biology, transcriptomics, and functional genomics are beginning to address these gaps, while emerging therapeutic approaches-such as small-molecule modulators and antibodies-highlight the potential of FZDs as drug targets. This review synthesizes current insights into FZD receptor biology, examines ongoing controversies, and outlines promising directions for future research and therapeutic development.
Collapse
Affiliation(s)
| | | | | | - Kevin Pruitt
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
4
|
Fang D, Zhou L, Zheng B. Research Progress on the Immunological Correlation Between Papillary Thyroid Carcinoma and Hashimoto's Thyroiditis. J Immunol Res 2025; 2025:7192808. [PMID: 40313970 PMCID: PMC12043394 DOI: 10.1155/jimr/7192808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
In recent years, a growing body of evidence has suggested a correlation between Hashimoto's thyroiditis (HT) and the onset and progression of papillary thyroid carcinoma (PTC). However, the mechanism underlying the relationship between HT and PTC remains incompletely understood. This review discusses the literature on the correlation between PTC and HT and summarizes the research concerning the immunological interplay between these two conditions. It also delves into tumor-associated cells (such as CD8+ T cells), tumor-associated macrophages (TAMs), regulatory T cells (Tregs), and cancer-associated fibroblasts (CAFs), alongside other tumor-associated factors, including interleukins (ILs), interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and hypoxia-inducible factor-1 (HIF-1), highlighting their roles in the interaction between PTC and HT. We also explore the strategic direction of immunotherapy in thyroid malignancies, particularly PTC with HT, and propose novel targeted immunotherapies for advanced thyroid cancer.
Collapse
Affiliation(s)
- Digui Fang
- Department of Thyroid and Parathyroid Surgery, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Limei Zhou
- Department of Thyroid and Parathyroid Surgery, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Biao Zheng
- Department of Thyroid and Parathyroid Surgery, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
5
|
Na W, Zeng W, Song K, Wang Y, Wang L, Zhao Z, Jin L, Zhu P, Liang W. PKM2, the "K+ sink" in the tumor interstitial fluid. Protein Cell 2025; 16:303-308. [PMID: 38912824 PMCID: PMC12053478 DOI: 10.1093/procel/pwae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/13/2024] [Accepted: 06/02/2024] [Indexed: 06/25/2024] Open
Affiliation(s)
- Wenjing Na
- Key Laboratory of Biomacromolecules (CAS), Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100864, China
| | - Wenfeng Zeng
- Key Laboratory of Biomacromolecules (CAS), Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100864, China
| | - Kai Song
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100864, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China
| | - Youwang Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100864, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China
| | - Luoyang Wang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Ziran Zhao
- Thoracic Surgery Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lingtao Jin
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Ping Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100864, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Liang
- Key Laboratory of Biomacromolecules (CAS), Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100864, China
| |
Collapse
|
6
|
Liu Y, Gao M, Yan Y, Wang X, Dong Z, Cheng L, Xu Y. Immunology in Osseointegration After Implantation. J Biomed Mater Res B Appl Biomater 2025; 113:e35566. [PMID: 40130467 DOI: 10.1002/jbm.b.35566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/26/2025]
Abstract
Bone tissue is renowned for its regenerative capabilities, yet handling extensive defects and complex fractures presents considerable medical challenges. Osteoimmunology, studying the complex mechanism of the mutual influence within the range of immunity and skeletal systems, has highlighted the indispensable function of immune reactions in the process of bone integration. This procedure, primarily immune-driven, fosters new bone formation around implants instead of relying solely on osteogenic mechanisms. Traditionally, implant research has emphasized bone bonding and osteoinduction, often overlooking the significant influence of immune interactions. Implants pose risks including donor site morbidity, decreased bioactivity, and pathogen transmission risks. To mitigate these, implant surfaces are modified through altering local factors such as electrical fields and applying physical treatments to change roughness, hydrophilicity, and nanotopography. These modifications aim to regulate immune reactions at the surface of the bone implant, improving osseointegration and the repair of bone. This review examines the types of immune cells in osseointegration, especially the pivotal function that macrophages serve in the regeneration of bone tissue, and investigates key implant features-morphology, wettability, cytokine interaction, and metal ion and bioactive molecule adsorption-that impact immune responses. These insights underscore the immune system's importance in bone repair and advance osteoimmunology as essential for developing strategies to control bone immune responses, enhancing implant integration and bone regeneration.
Collapse
Affiliation(s)
- Yuyang Liu
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Min Gao
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Yikun Yan
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Xue Wang
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Zhihong Dong
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Lijia Cheng
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Yaji Xu
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| |
Collapse
|
7
|
Wang Y, Xu N, Ndzie Noah ML, Chen L, Zhan X. Pyruvate Kinase M1/2 Proteoformics for Accurate Insights into Energy Metabolism Abnormity to Promote the Overall Management of Ovarian Cancer Towards Predictive, Preventive, and Personalized Medicine Approaches. Metabolites 2025; 15:203. [PMID: 40137167 PMCID: PMC11944880 DOI: 10.3390/metabo15030203] [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: 02/02/2025] [Revised: 03/01/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Ovarian cancer (OC) is a global health problem that frequently presents at advanced stages, is predisposed to recurrence, readily develops resistance to platinum-based drugs, and has a low survival rate. Predictive, preventive, and personalized medicine (PPPM/3PM) offers an integrated solution with the use of genetic, proteomic, and metabolic biomarkers to identify high-risk individuals for early detection. Metabolic reprogramming is one of the key strategies employed by tumor cells to adapt to the microenvironment and support unlimited proliferation. Pyruvate kinases M1 and M2 (PKM1/2) are encoded by the PKM gene, a pivotal enzyme in the last step of the glycolytic pathway, which is at the crossroads of aerobic oxidation and the Warburg effect to serve as a potential regulator of glucose metabolism and influence cellular energy production and metabolic reprogramming. Commonly, the ratio of PKM1-to-PKM2 is changed in tumors compared to normal controls, and PKM2 is highly expressed in OC to induce a high glycolysis rate and participate in the malignant invasion and metastatic characteristics of cancer cells with epithelial/mesenchymal transition (EMT). PKM2 inhibitors suppress the migration and growth of OC cells by interfering with the Warburg effect. Proteoforms are the final structural and functional forms of a gene/protein, and the canonical protein PKM contains all proteoforms encoded by the same PKM gene. The complexity of PKM can be elucidated by proteoformics. The OC-specific PKM proteoform might represent a specific target for therapeutic interventions against OC. In the framework of PPPM/3PM, the OC-specific PKM proteoform might be the early warning and prognosis biomarker. It is important to clarify the molecular mechanisms of PKM proteoforms in cancer metabolism. This review analyzes the expression, function, and molecular mechanisms of PKM proteoforms in OC, which help identify specific biomarkers for OC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China
- Department of Gynecology, Gaotang County Medical Center, Liaocheng 252800, China
| | - Nuo Xu
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
| | - Marie Louise Ndzie Noah
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
| | - Liang Chen
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China
| | - Xianquan Zhan
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China
| |
Collapse
|
8
|
Song S, Li F, Zhao B, Zhou M, Wang X. Ultraviolet Light Causes Skin Cell Senescence: From Mechanism to Prevention Principle. Adv Biol (Weinh) 2025; 9:e2400090. [PMID: 39364703 DOI: 10.1002/adbi.202400090] [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: 02/18/2024] [Revised: 08/23/2024] [Indexed: 10/05/2024]
Abstract
The skin is an effective protective barrier that significantly protects the body from damage caused by external environmental factors. Furthermore, skin condition significantly affects external beauty. In today's era, which is of material and spiritual prosperity, there is growing attention on skincare and wellness. Ultraviolet radiation is one of the most common external factors that lead to conditions like sunburn, skin cancer, and skin aging. In this review, several mechanisms of UV-induced skin cell senescence are discussed, including DNA damage, oxidative stress, inflammatory response, and mitochondrial dysfunction, which have their own characteristics and mutual effects. As an illustration, mitochondrial dysfunction triggers electron evasion and the generation of more reactive oxygen species, leading to oxidative stress and the activation of the NLRP3 inflammasome, which in turn causes mitochondrial DNA (mt DNA) damage. Based on the current mechanism, suitable prevention and treatment strategies are proposed from sunscreen, dietary, and experimental medications respectively, aimed at slowing down skin cell aging and providing protection from ultraviolet radiation. The effects of ultraviolet rays on skin is summarized, offering insights and directions for future studies on mechanism of skin cell senescence, with an anticipation of discovering more effective prevention and cure methods.
Collapse
Affiliation(s)
- Shujia Song
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Fuxing Li
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Bingxiang Zhao
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Min Zhou
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Xiaobo Wang
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| |
Collapse
|
9
|
Dalpati N, Rai SK, Sharma P, Sarangi PP. Integrins and integrin-driven secretory pathways as multi-dimensional regulators of tumor-associated macrophage recruitment and reprogramming in tumor microenvironment. Matrix Biol 2025; 135:55-69. [PMID: 39645091 DOI: 10.1016/j.matbio.2024.12.003] [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: 06/30/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Integrins, a group of transmembrane receptors, play a crucial role in mediating the interactions between cells and extracellular matrix (ECM) proteins. The intracellular signaling initiated by these cell-matrix interactions in leukocytes mediates many essential cellular processes such as survival, migration, metabolism, and other immunological functions. Macrophages, as phagocytes, participate in both proinflammatory and anti-inflammatory processes, including progression. Numerous reports have shown that the integrin-regulated secretome, comprising cytokines, chemokines, growth factors, proteases, and other bioactive molecules, is a crucial modulator of macrophage functions in tumors, significantly influencing macrophage programming and reprogramming within the tumor microenvironment (TME) in addition to driving their step-by-step entry process into tumor tissue spaces. Importantly, studies have demonstrated a pivotal role for integrin receptor-mediated secretome and associated signaling pathways in functional reprogramming from anti-tumorigenic to pro-tumorigenic phenotype in tumor-associated macrophages (TAMs). In this comprehensive review, we have provided an in-depth analysis of the latest findings of various key pathways, mediators, and signaling cascades associated with integrin-driven polarization of macrophages in tumors. This manuscript will provide an updated understanding of the modulation of inflammatory monocytes/ macrophages and TAMs by integrin-driven secretory pathways in various functions such as migration, differentiation, and their role in tumor progression, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Prerna Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
10
|
Xia Y, Huang C, Zhong M, Zhong H, Ruan R, Xiong J, Yao Y, Zhou J, Deng J. Targeting HGF/c-MET signaling to regulate the tumor microenvironment: Implications for counteracting tumor immune evasion. Cell Commun Signal 2025; 23:46. [PMID: 39856684 PMCID: PMC11762533 DOI: 10.1186/s12964-025-02033-1] [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: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor (HGF) along with its receptor (c-MET) are crucial in preserving standard cellular physiological activities, and imbalances in the c-MET signaling pathway can lead to the development and advancement of tumors. It has been extensively demonstrated that immune checkpoint inhibitors (ICIs) can result in prolonged remission in certain patients. Nevertheless, numerous preclinical studies have shown that MET imbalance hinders the effectiveness of anti-PD-1/PD-L1 treatments through various mechanisms. Consequently, clarifying the link between the c-MET signaling pathway and the tumor microenvironment (TME), as well as uncovering the effects of anti-MET treatment on ICI therapy, is crucial for enhancing the outlook for tumor patients. In this review, we examine the impact of abnormal activation of the HGF/c-MET signaling pathway on the control of the TME and the processes governing PD-L1 expression in cancer cells. The review thoroughly examines both clinical and practical evidence regarding the use of c-MET inhibitors alongside PD-1/PD-L1 inhibitors, emphasizing that focusing on c-MET with immunotherapy enhances the effectiveness of treating MET tumors exhibiting elevated PD-L1 expression.
Collapse
Affiliation(s)
- Yang Xia
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Min Zhong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jing Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
11
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 PMCID: PMC11817814 DOI: 10.3390/cells14030170] [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: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A. Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia (V.G.T.)
| | | | | | | | | |
Collapse
|
12
|
Wu L, Liu C, Hu W. Comprehensive investigation of matrix metalloproteinases in skin cutaneous melanoma: diagnostic, prognostic, and therapeutic insights. Sci Rep 2025; 15:2152. [PMID: 39820824 PMCID: PMC11739484 DOI: 10.1038/s41598-025-85887-2] [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: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
The dysregulation of matrix metalloproteinases (MMPs) in skin cutaneous melanoma (SKCM) represents a critical aspect of tumorigenesis. In this study, we investigated the diagnostic, prognostic, and therapeutic aspects of the MMPs in SKCM. Thirteen SKCM cell lines and seven normal skin cell lines were cultured under standard conditions for experimental analyses. RNA and DNA were extracted, followed by RT-qPCR to assess MMP expression and promoter methylation analysis to determine methylation levels. Functional assays, including cell proliferation, colony formation, and wound healing, were conducted post-MMP7 knockdown using siRNA in A375 cells. Databases like GEPIA2, HPA, MEXPRESS, and miRNet were employed for expression, survival, methylation, and miRNA-mRNA network analyses. We investigated the expression and promoter methylation landscape of MMPs in SKCM cell lines, revealing significant (p-value < 0.05) up-regulation of MMP1, MMP7, MMP9, MMP10, MMP11, MMP12, MMP13, MMP14, and MMP25, alongside down-regulation of MMP2, MMP3, and MMP21. Furthermore, our analysis demonstrated a significant (p-value < 0.05) inverse correlation between MMP expression levels and promoter methylation status, suggesting a potential regulatory role of DNA methylation in MMP dysregulation. Notably, MMP7, MMP11, and MMP14 exhibited significant (p-value < 0.05) associations with the overall survival of SKCM patients, emphasizing their prognostic significance. Additionally, Receiver operating characteristic (ROC) curve analysis highlighted the significant (p-value < 0.05) diagnostic potential of MMP7, MMP11, and MMP14 in distinguishing SKCM from normal individuals. Subsequent validation across multiple cohorts confirmed significant (p-value < 0.05) elevated MMP expression levels in SKCM tissues, particularly in advanced disease stages, further emphasizing their role in tumor progression. Furthermore, we elucidated potential regulatory pathways involving miR-22-3p, which targets MMP7, MMP11, and MMP14 genes in SKCM. Our findings also revealed associations between MMP expression and immune modulation, drug sensitivity, and functional states of SKCM cells. Lastly, MMP7 knockdown in A375 cells significantly significant (p-value < 0.05) impacted several characteristics, including cell proliferation, colony formation, and wound healing. Our findings highlight the diagnostic, prognostic, and therapeutic potential of MMP7, MMP11, and MMP14 in SKCM. These MMPs could serve as biomarkers for early detection and targets for therapy. Future efforts should focus on preclinical and clinical validation to translate these insights into personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lingxia Wu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China
| | - Chenxiaoxiao Liu
- The First Clinical Institute, Zunyi Medical University, Zunyi, 520300, Guizhou, China
| | - Weicai Hu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China.
| |
Collapse
|
13
|
Jang B, Amirshaghaghi A, Choi J, Miller J, Issadore DA, Busch TM, Cheng Z, Tsourkas A. Enhanced Accumulation and Penetration of Magnetic Nanoclusters in Tumors Using an 8-Magnet Halbach Array Leads to Improved Cancer Treatment. ACS NANO 2025; 19:1794-1808. [PMID: 39749910 PMCID: PMC11735326 DOI: 10.1021/acsnano.4c16600] [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] [Indexed: 01/04/2025]
Abstract
Nanoparticles have gained attention as drug delivery vehicles for cancer treatment, but often struggle with poor tumor accumulation and penetration. Single external magnets can enhance magnetic nanoparticle delivery but are limited to superficial tumors due to the rapid decline in the magnetic field strength with distance. We previously showed that a 2-magnet device could extend targeting to greater tissue depths. Here, we improve on this approach by constructing an 8-magnet device arranged in an annular Halbach array, which facilitates radial outward movement of magnetic nanoparticles from the bore's center. Using chlorin e6-coated magnetic nanoclusters (Ce6 clusters) with densely packed cobalt-doped superparamagnetic iron oxide nanoparticles, we demonstrated nearly a 7-fold improvement in nanoparticle movement through a porous matrix compared to the 2-magnet approach. This resulted in enhanced magnetic resonance contrast, accumulation, and penetration of Ce6 clusters into 4T1 triple-negative breast tumors in mice, leading to improved photodynamic therapy and highlighting the potential therapeutic application of the 8-magnet device.
Collapse
Affiliation(s)
- Bian Jang
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 435 Skirkanich Hall, Philadelphia, PA 19104, United States
| | - Ahmad Amirshaghaghi
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 435 Skirkanich Hall, Philadelphia, PA 19104, United States
| | - Jeongmoon Choi
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Joann Miller
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, United States
| | - David A. Issadore
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 435 Skirkanich Hall, Philadelphia, PA 19104, United States
| | - Theresa M. Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, United States
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 435 Skirkanich Hall, Philadelphia, PA 19104, United States
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 435 Skirkanich Hall, Philadelphia, PA 19104, United States
| |
Collapse
|
14
|
Serratì S, Zerlotin R, Manganelli M, Di Fonte R, Dicarlo M, Oranger A, Colaianni G, Porcelli L, Azzariti A, Guida S, Grano M, Colucci SC, Guida G. Irisin and Metastatic Melanoma: Selective Anti-Invasiveness Activity in BRAF Wild-Type Cells. Int J Mol Sci 2025; 26:652. [PMID: 39859367 PMCID: PMC11765811 DOI: 10.3390/ijms26020652] [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: 12/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Irisin is a newly discovered 12 kDa messenger protein involved in energy metabolism. Irisin affects signaling pathways in several types of cancer; however, the role of irisin in metastatic melanoma (MM) has not been described yet. We explored the biological effects of irisin in in vitro models of MM cells (HBLwt/wt, LND1wt/wt, Hmel1V600K/wt and M3V600E/V600E) capable of the oncogenic activation of BRAF. We treated MM cells with different concentrations of r-irisin (10 nM, 25 nM, 50 nM, 100 nM) for 24 h-48 h. An MTT assay highlighted that r-irisin did not affect the proliferation of MM cells. We subsequently treated MM cells with 10 nM r-irisin, corresponding to the dose exhibiting biological activity in vitro. Irisin reduced the invasive ability of only LND1wt/wt (p < 0.05), which highly expressed αv gene levels, but did not affect the invasion of BRAFmut cells. Gelatin zymography analysis showed a reduction in the enzymatic activity of MMP-2 and MMP-9 in BRAFwt/wt cells treated with 10 nM r-irisin. Moreover, gene expression analysis (qPCR) of MMP-2 and MMP-9 and of the fibrinolytic system (uPAR, uPA and PAI-1) highlighted a crucial role of 10 nM r-irisin treatment in the inhibition of pro-invasive systems in BRAFwt/wt. In conclusion, our results may suggest a possible differential role of irisin in melanoma cells.
Collapse
Affiliation(s)
- Simona Serratì
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Roberta Zerlotin
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Michele Manganelli
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy; (M.M.); (S.C.C.)
| | - Roberta Di Fonte
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Manuela Dicarlo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Angela Oranger
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Letizia Porcelli
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Amalia Azzariti
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Stefania Guida
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy;
- Dermatology Clinic, IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Silvia Concetta Colucci
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy; (M.M.); (S.C.C.)
| | - Gabriella Guida
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy; (M.M.); (S.C.C.)
| |
Collapse
|
15
|
Hernandez K, Nguyen CH, Rijal G. Asporin increases the extracellular matrix cross-links and inhibits the cancer cell migration. Tumour Biol 2025; 47:10104283241313441. [PMID: 40099523 DOI: 10.1177/10104283241313441] [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] [Indexed: 03/20/2025] Open
Abstract
BackgroundMigrating strategies of the triple-negative breast cancer (TNBC) together with its role in the establishment of tumor microenvironment (TME), supporting metastasis, have been extensively studied. Extracellular matrix (ECM) is a major player for the TME, establishing the 3D spatial networks with interconnected pores necessary for the mechano-physiological function of the cells. Certain collagen aligners and cross-linkers which are necessary for the formation and the stabilization of ECM networks, however, have not been studied either in normal or in abnormal tissues. Complexities in cell-cell and cell-matrix interactions, and different in types and ratios of ECM proteins in a TME challenge to reveal the precise function of a particular protein that is exhibited by special cells and if specifically present in insignificant amount. Cancer-associated fibroblasts (CAFs) predominantly occupy the major stroma of a solid tumor where they deposit extracellular proteins in the excessive amount compared to other tumor-associated cells. For example, the TNBC tumor itself is positive for asporin (ASPN) since CAFs are major ASPN exhibitors. However, the TNBC cells express it insignificantly.ObjectiveThe increase in ECM and its networks suppresses the metastasis.MethodsHere, we studied the expression of collagen type I and ASPN in CAFS and MDA-MB-231 (MM231), and evaluated the role of ASPN in collagen alignment and crosslinking.ResultsTNBC cells have an insignificant expression of ASPN and scanty collagen fibers, some of which aggregate to form the stiff deranged fibers, forming large-size pores in ECM of cancer-cell-dominant outer core of TNBC that support cancer cell invasion and metastasis. Exogenous ASPN and fibroblast-ASPN supported for the collagen alignment and crosslinking that established the small-size pores in the ECM, inhibiting the cancer cell invasion.ConclusionsThe collagen aligner and the cross-linker, ASPN increases the ECM networks and decreases the migration, and this preliminary study provides the hope that ASPN might be used as an anti-metastatic drug after its confirmation through extensive studies in animal, and positive outcomes through preclinical trials.
Collapse
Affiliation(s)
- Kimberly Hernandez
- Department of Medical Laboratory Sciences, Public Health and Nutrition Science, Tarleton State University, A Member of Texas A & M University System, Fort Worth, TX, USA
| | - Caitlin H Nguyen
- Department of Medical Laboratory Sciences, Public Health and Nutrition Science, Tarleton State University, A Member of Texas A & M University System, Fort Worth, TX, USA
| | - Girdhari Rijal
- Department of Medical Laboratory Sciences, Public Health and Nutrition Science, Tarleton State University, A Member of Texas A & M University System, Fort Worth, TX, USA
| |
Collapse
|
16
|
Wu Y, Jiang X, Yu Z, Xing Z, Ma Y, Qing H. Mechanisms of Anti-PD Therapy Resistance in Digestive System Neoplasms. Recent Pat Anticancer Drug Discov 2025; 20:1-25. [PMID: 38305306 PMCID: PMC11865675 DOI: 10.2174/0115748928269276231120103256] [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: 06/27/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 02/03/2024]
Abstract
Digestive system neoplasms are highly heterogeneous and exhibit complex resistance mechanisms that render anti-programmed cell death protein (PD) therapies poorly effective. The tumor microenvironment (TME) plays a pivotal role in tumor development, apart from supplying energy for tumor proliferation and impeding the body's anti-tumor immune response, the TME actively facilitates tumor progression and immune escape via diverse pathways, which include the modulation of heritable gene expression alterations and the intricate interplay with the gut microbiota. In this review, we aim to elucidate the mechanisms underlying drug resistance in digestive tumors, focusing on immune-mediated resistance, microbial crosstalk, metabolism, and epigenetics. We will highlight the unique characteristics of each digestive tumor and emphasize the significance of the tumor immune microenvironment (TIME). Furthermore, we will discuss the current therapeutic strategies that hold promise for combination with cancer immune normalization therapies. This review aims to provide a thorough understanding of the resistance mechanisms in digestive tumors and offer insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuxia Wu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zongrui Xing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yong Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Huiguo Qing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
17
|
Xi Y, Collins LB, Bai H, Biehl A, Mora-Navarro C, Freytes D, Islam Williams T. A Multi-enzyme Protocol Improves Total Proteome Coverage in Extracellular Matrix. Methods Mol Biol 2025; 2884:179-191. [PMID: 39716004 DOI: 10.1007/978-1-0716-4298-6_12] [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] [Indexed: 12/25/2024]
Abstract
Extracellular matrix (ECM) from decellularized mammalian tissues has been used in many therapeutic applications. The tissue-specific composition of the ECM is critically associated with therapeutic performance. However, ECM translation needs to be improved because of the complex composition and limited understanding of ECM repairing mechanisms due partly to incomplete proteomic interrogation of ECM samples. In this chapter, we describe a multi-enzyme, bottom-up proteomics workflow employing trypsin, Lys-C, collagenase, and elastase to enhance the digestion of ECM and increase total protein coverage. The outcomes from the reported approach, in a standardized manner, enable users to pinpoint changes in the ECM composition, thereby facilitating the establishment of mechanistic correlations between ECM composition and its effects.
Collapse
Affiliation(s)
- Ying Xi
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | - Leonard B Collins
- Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, NC, USA
| | - Hongxia Bai
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | - Andreea Biehl
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Camilo Mora-Navarro
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez, Mayaguez, PR, USA
| | - Donald Freytes
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Taufika Islam Williams
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA.
- Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
18
|
Lv K, He T. Cancer-associated fibroblasts: heterogeneity, tumorigenicity and therapeutic targets. MOLECULAR BIOMEDICINE 2024; 5:70. [PMID: 39680287 PMCID: PMC11649616 DOI: 10.1186/s43556-024-00233-8] [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: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Cancer, characterized by its immune evasion, active metabolism, and heightened proliferation, comprises both stroma and cells. Although the research has always focused on parenchymal cells, the non-parenchymal components must not be overlooked. Targeting cancer parenchymal cells has proven to be a formidable challenge, yielding limited success on a broad scale. The tumor microenvironment(TME), a critical niche for cancer cell survival, presents a novel way for cancer treatment. Cancer-associated fibroblast (CAF), as a main component of TME, is a dynamically evolving, dual-functioning stromal cell. Furthermore, their biological activities span the entire spectrum of tumor development, metastasis, drug resistance, and prognosis. A thorough understanding of CAFs functions and therapeutic advances holds significant clinical implications. In this review, we underscore the heterogeneity of CAFs by elaborating on their origins, types and function. Most importantly, by elucidating the direct or indirect crosstalk between CAFs and immune cells, the extracellular matrix, and cancer cells, we emphasize the tumorigenicity of CAFs in cancer. Finally, we highlight the challenges encountered in the exploration of CAFs and list targeted therapies for CAF, which have implications for clinical treatment.
Collapse
Affiliation(s)
- Keke Lv
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China
| | - Tianlin He
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
19
|
Liu H, Wang X, Liu L, Yan B, Qiu F, Zhou B. Targeting liposarcoma: unveiling molecular pathways and therapeutic opportunities. Front Oncol 2024; 14:1484027. [PMID: 39723387 PMCID: PMC11668776 DOI: 10.3389/fonc.2024.1484027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
In recent years, an increasing number of studies have utilized molecular biology techniques to reveal important molecular heterogeneity among different subtypes of liposarcoma. Each subtype exhibits distinct genetic patterns and molecular pathways, which may serve as important targets for molecular therapy. In the present review, we focus on the molecular characteristics, molecular diagnostics, driver genes, and molecular mechanisms of liposarcoma. We also discuss the clinical research progress of related targeted therapies, with an aim to provide a reference and crucial insights for colleagues in the field.
Collapse
Affiliation(s)
- Hongliang Liu
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xi Wang
- Department of Oncology, Women and Children’s Hospital Affiliated to Qingdao University, Qingdao, China
| | - Lingyan Liu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Bingsong Yan
- Department of Hepatobiliary Surgery, Women and Children’s Hospital Affiliated to Qingdao University, Qingdao, China
| | - Fabo Qiu
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Zhang Y, Dong X, Zhang Y, Chen Z, Zhou G, Chen N, Shen W, Yang K, Pei P. Biomaterials to regulate tumor extracellular matrix in immunotherapy. J Control Release 2024; 376:149-166. [PMID: 39389365 DOI: 10.1016/j.jconrel.2024.10.010] [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: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The tumor extracellular matrix (ECM) provides physical support and influences tumor development, metastasis, and the tumor microenvironment, creating barriers to immune drug delivery and cell infiltration. Therefore, modulating or degrading the ECM is of significant importance to enhance the efficacy of tumor immunotherapy. This manuscript initially summarizes the main strategies and mechanisms of biomaterials in modulating various components of the ECM, including collagen, fibronectin, hyaluronic acid, and in remodeling the ECM. Subsequently, it discusses the benefits of biomaterials for immunotherapy following ECM modulation, such as promoting the infiltration of drugs and immune cells, regulating immune cell function, and alleviating the immunosuppressive microenvironment. The manuscript also briefly introduces the application of biomaterials that utilize and mimic the ECM for tumor immunotherapy. Finally, it addresses the current challenges and future directions in this field, providing a comprehensive overview of the potential and innovation in leveraging biomaterials to enhance cancer treatment outcomes. Our work will offer a comprehensive overview of ECM modulation strategies and their application in biomaterials to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Yujie Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xuexue Dong
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zetong Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Ni Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Jiangsu, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province 230022, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| |
Collapse
|
21
|
Lee CJ, Jang TY, Jeon SE, Yun HJ, Cho YH, Lim DY, Nam JS. The dysadherin/MMP9 axis modifies the extracellular matrix to accelerate colorectal cancer progression. Nat Commun 2024; 15:10422. [PMID: 39613801 PMCID: PMC11607440 DOI: 10.1038/s41467-024-54920-9] [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: 02/12/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024] Open
Abstract
The dynamic alteration of the tumor microenvironment (TME) serves as a driving force behind the progression and metastasis of colorectal cancer (CRC). Within the intricate TME, a pivotal player is the extracellular matrix (ECM), where modifications in components, degradation, and stiffness are considered critical factors in tumor development. In this study, we find that the membrane glycoprotein dysadherin directly targets matrix metalloprotease 9 (MMP9), initiating ECM remodeling within the TME and amplifying cancer progression. Mechanistically, the dysadherin/MMP9 axis not only enhances CRC cell invasiveness and ECM proteolytic activity but also activates cancer-associated fibroblasts, orchestrating the restructuring of the ECM through the synthesis of its components in human CRC cells, patient samples, and mouse models. Notably, disruption of ECM reorganization by dysadherin knockout results in a discernible reduction in the immunosuppressive and proangiogenic milieu in a humanized mouse model. Intriguingly, these effects are reversed upon the overexpression of MMP9, highlighting the intricate and pivotal role of the dysadherin/MMP9 axis in shaping the development of a malignant TME. Therefore, our findings not only highlight that dysadherin contributes to CRC progression by influencing the TME through ECM remodeling but also suggest that dysadherin may be a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Choong-Jae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Tae-Young Jang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - So-El Jeon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyeon-Ji Yun
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Yeong-Hoon Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Da-Ye Lim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jeong-Seok Nam
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
22
|
Setayesh T, Hu Y, Vaziri F, Wei D, Wan YJY. The spatial impact of a Western diet in enriching Galectin-1-regulated Rho, ECM, and SASP signaling in a novel MASH-HCC mouse model. Biomark Res 2024; 12:122. [PMID: 39402682 PMCID: PMC11476289 DOI: 10.1186/s40364-024-00660-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) arising from metabolic dysfunction-associated steatohepatitis (MASH) presents a significant clinical challenge, particularly given the prevalence of the Western diet (WD). The influence of diet on the tumor microenvironment remains poorly understood. Galectin-1 (Gal-1) is a biomarker for HCC and has a crucial role in liver carcinogenesis. Our previous studies demonstrated that silencing Gal-1 effectively treats mouse HCC. However, the impacts of a WD on Gal-1 signaling on MASH to HCC progression are unknown, and this study addresses these knowledge gaps. METHODS We developed a novel MASH-HCC mouse model. Using spatial transcriptomics and multiplex immunohistochemistry (IHC), we studied the effects of a WD on the liver and tumor microenvironment. By modulating Gal-1 expression through silencing and overexpression, we explored the location-specific impacts of WD on Gal-1 signaling. RESULTS Pathways such as Rho signaling, extracellular matrix (ECM) remodeling, and senescence-associated secretory phenotypes (SASP) were prominently activated in WD-induced metabolic dysfunction-associated fatty liver disease (MAFLD) and MASH-HCC, compared to healthy livers controls. Furthermore, Rho GTPase effectors, ECM remodeling, neutrophil degranulation, cellular stress, and cell cycle pathways were consistently enriched in human and mouse MASH-HCC. Spatially, these pathways were enriched in the tumor and tumor margins of mouse MASH-HCC. Additionally, there was a notable increase in CD11c and PD-L1-positive cells from non-tumor tissues to the tumor margin and inside the tumor of MASH-HCC, suggesting compromised immune surveillance due to WD intake. Moreover, MASH-HCC exhibited significant Gal-1 induction in N-Cadherin-positive cells, indicating enhanced epithelial-to-mesenchymal transition (EMT). Modulating Gal-1 expression in MASH-HCC further established its specific roles in regulating Rho signaling and SASP in the tumor margin and non-tumor tissues in MASH-HCC. CONCLUSION WD intake significantly influences vital cellular processes involved in Gal-1-mediated signaling, including Rho signaling and ECM remodeling, in the tumor microenvironment, thereby contributing to the development of MASH-HCC.
Collapse
Affiliation(s)
- Tahereh Setayesh
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Ying Hu
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Farzam Vaziri
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Dongguang Wei
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA.
| |
Collapse
|
23
|
Najafzadeh M, Sajjadi SM, Kharazi S, Karimifard F, Safarpour H, Kharazinejad E. Interactions between cancer and stroma mediated by extracellular vesicles. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:114. [DOI: 10.1186/s43042-024-00582-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/13/2024] [Indexed: 01/05/2025] Open
Abstract
AbstractExtracellular vehicles (EVs) are small membrane-bound particles that are released by both cancer and stromal cells. These vesicles have emerged as key mediators of intercellular communication within the tumor microenvironment. In particular, EVs have been shown to play a critical role in facilitating the interactions between cancer cells and the surrounding stroma. Through the transfer of various bioactive molecules, including proteins, lipids, and nucleic acids, EVs are able to modulate the behavior of recipient cells and promote tumorigenesis. Additionally, EVs can also contribute to the development of drug resistance and immune evasion, further highlighting their importance in cancer progression. This review will summarize the current knowledge regarding EV-mediated interactions between cancer and stromal cells, and discuss their implications for cancer diagnosis and therapy.
Collapse
|
24
|
Stewart DC, Brisson BK, Dekky B, Berger AC, Yen W, Mauldin EA, Loebel C, Gillette D, Assenmacher CA, Quincey C, Stefanovski D, Cristofanilli M, Cukierman E, Burdick JA, Borges VF, Volk SW. Prognostic and therapeutic implications of tumor-restrictive type III collagen in the breast cancer microenvironment. NPJ Breast Cancer 2024; 10:86. [PMID: 39358397 PMCID: PMC11447064 DOI: 10.1038/s41523-024-00690-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Collagen plays a critical role in regulating breast cancer progression and therapeutic resistance. An improved understanding of both the features and drivers of tumor-permissive and -restrictive collagen matrices are critical to improve prognostication and develop more effective therapeutic strategies. In this study, using a combination of in vitro, in vivo and bioinformatic experiments, we show that type III collagen (Col3) plays a tumor-restrictive role in human breast cancer. We demonstrate that Col3-deficient, human fibroblasts produce tumor-permissive collagen matrices that drive cell proliferation and suppress apoptosis in non-invasive and invasive breast cancer cell lines. In human triple-negative breast cancer biopsy samples, we demonstrate elevated deposition of Col3 relative to type I collagen (Col1) in non-invasive compared to invasive regions. Similarly, bioinformatics analysis of over 1000 breast cancer patient biopsies from The Cancer Genome Atlas BRCA cohort revealed that patients with higher Col3:Col1 bulk tumor expression had improved overall, disease-free, and progression-free survival relative to those with higher Col1:Col3 expression. Using an established 3D culture model, we show that Col3 increases spheroid formation and induces the formation of lumen-like structures that resemble non-neoplastic mammary acini. Finally, our in vivo study shows co-injection of murine breast cancer cells (4T1) with rhCol3-supplemented hydrogels limits tumor growth and decreases pulmonary metastatic burden compared to controls. Taken together, these data collectively support a tumor-suppressive role for Col3 in human breast cancer and suggest that strategies that increase Col3 may provide a safe and effective therapeutic modality to limit recurrence in breast cancer patients.
Collapse
Affiliation(s)
- Daniel C Stewart
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Becky K Brisson
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bassil Dekky
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashton C Berger
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William Yen
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth A Mauldin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Claudia Loebel
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Deborah Gillette
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Corisa Quincey
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Darko Stefanovski
- Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, The Martin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Jason A Burdick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Virginia F Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Susan W Volk
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Tobi A, Haugas M, Rabi K, Sethi J, Põšnograjeva K, Paiste P, Jagomäe T, Pleiko K, Lingasamy P, Teesalu T. Protease-activated CendR peptides targeting tenascin-C: mitigating off-target tissue accumulation. Drug Deliv Transl Res 2024; 14:2945-2961. [PMID: 39012578 PMCID: PMC11384632 DOI: 10.1007/s13346-024-01670-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
To achieve precision and selectivity, anticancer compounds and nanoparticles (NPs) can be targeted with affinity ligands that engage with malignancy-associated molecules in the blood vessels. While tumor-penetrating C-end Rule (CendR) peptides hold promise for precision tumor delivery, C-terminally exposed CendR peptides can accumulate undesirably in non-malignant tissues expressing neuropilin-1 (NRP-1), such as the lungs. One example of such promiscuous peptides is PL3 (sequence: AGRGRLVR), a peptide that engages with NRP-1 through its C-terminal CendR element, RLVR.Here, we report the development of PL3 derivatives that bind to NRP-1 only after proteolytic processing by urokinase-type plasminogen activator (uPA), while maintaining binding to the other receptor of the peptide, the C-domain of tenascin-C (TNC-C). Through a rational design approach and screening of a uPA-treated peptide-phage library (PL3 peptide followed by four random amino acids) on the recombinant NRP-1, derivatives of the PL3 peptide capable of binding to NRP-1 only post-uPA processing were successfully identified. In vitro cleavage, binding, and internalization assays, along with in vivo biodistribution studies in orthotopic glioblastoma-bearing mice, confirmed the efficacy of two novel peptides, PL3uCendR (AGRGRLVR↓SAGGSVA) and SKLG (AGRGRLVR↓SKLG), which exhibit uPA-dependent binding to NRP-1, reducing off-target binding to healthy NRP-1-expressing tissues. Our study not only unveils novel uPA-dependent TNC-C targeting CendR peptides but also introduces a broader paradigm and establishes a technology for screening proteolytically activated tumor-penetrating peptides.
Collapse
Affiliation(s)
- Allan Tobi
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia
| | - Maarja Haugas
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia
| | - Kristina Rabi
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia
| | - Jhalak Sethi
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia
| | - Kristina Põšnograjeva
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia
| | - Päärn Paiste
- Department of Geology, Institute of Ecology and Earth Sciences, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - Toomas Jagomäe
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia
| | - Karlis Pleiko
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia
| | - Prakash Lingasamy
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14B, 50411, Tartu, Estonia.
- Materials Research Laboratory, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
26
|
Doran BR, Moffitt LR, Wilson AL, Stephens AN, Bilandzic M. Leader Cells: Invade and Evade-The Frontline of Cancer Progression. Int J Mol Sci 2024; 25:10554. [PMID: 39408880 PMCID: PMC11476628 DOI: 10.3390/ijms251910554] [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: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Metastasis is the leading cause of cancer-related mortality; however, a complete understanding of the molecular programs driving the metastatic cascade is lacking. Metastasis is dependent on collective invasion-a developmental process exploited by many epithelial cancers to establish secondary tumours and promote widespread disease. The key drivers of collective invasion are "Leader Cells", a functionally distinct subpopulation of cells that direct migration, cellular contractility, and lead trailing or follower cells. While a significant body of research has focused on leader cell biology in the traditional context of collective invasion, the influence of metastasis-promoting leader cells is an emerging area of study. This review provides insights into the expanded role of leader cells, detailing emerging evidence on the hybrid epithelial-mesenchymal transition (EMT) state and the phenotypical plasticity exhibited by leader cells. Additionally, we explore the role of leader cells in chemotherapeutic resistance and immune evasion, highlighting their potential as effective and diverse targets for novel cancer therapies.
Collapse
Affiliation(s)
- Brittany R. Doran
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Laura R. Moffitt
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Amy L. Wilson
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Andrew N. Stephens
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| |
Collapse
|
27
|
Senigagliesi B, Geiss O, Valente S, Vondracek H, Cefarin N, Ceccone G, Calzolai L, Ballerini L, Parisse P, Casalis L. Substrate stiffness modulates extracellular vesicles' release in a triple-negative breast cancer model. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:553-568. [PMID: 39697626 PMCID: PMC11648499 DOI: 10.20517/evcna.2024.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 12/20/2024]
Abstract
Aim The microenvironment effect on the tumoral-derived Extracellular Vesicle release, which is of significant interest for biomedical applications, still represents a rather unexplored field. The aim of the present work is to investigate the interrelation between extracellular matrix (ECM) stiffness and the release of small EVs from cancer cells. Here, we focus on the interrelation between the ECM and small extracellular vesicles (sEVs), specifically investigating the unexplored aspect of the influence of ECM stiffness on the release of sEVs. Methods We used a well-studied metastatic Triple-Negative Breast Cancer (TNBC) cell line, MDA-MB-231, as a model to study the release of sEVs by cells cultured on substrates of different stiffness. We have grown MDA-MB-231 cells on two collagen-coated polydimethylsiloxane (PDMS) substrates at different stiffness (0.2 and 3.6 MPa), comparing them with a hard glass substrate as control, and then we isolated the respective sEVs by differential ultracentrifugation. After checking the cell growth conditions [vitality, morphology by immunofluorescence microscopy, stiffness by atomic force microscopy (AFM)], we took advantage of a multi-parametric approach based on complementary techniques (AFM, Nanoparticle Tracking Analysis, and asymmetric flow field flow fractionation with a multi-angle light scattering detector) to characterize the TNBC-derived sEV obtained in the different substrate conditions. Results We observe that soft substrates induce TNBC cell softening and rounding. This effect promotes the release of a high number of larger sEVs. Conclusion Here, we show the role of ECM physical properties in the regulation of sEV release in a TNBC model. While the molecular mechanisms regulating this effect need further investigation, our report represents a step toward an improved understanding of ECM-cell-sEVs crosstalk.
Collapse
Affiliation(s)
- Beatrice Senigagliesi
- Neuroscience Area, Scuola Internazionale Superiore di Studi Avanzati, Trieste 34136, Italy
- Nano-Innovation Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Trieste 34149, Italy
- Institute for Health and Consumer Protection, European Commission - Joint Research Centre, Ispra 21027, Italy
| | - Otmar Geiss
- Institute for Health and Consumer Protection, European Commission - Joint Research Centre, Ispra 21027, Italy
| | - Stefano Valente
- Institute for Health and Consumer Protection, European Commission - Joint Research Centre, Ispra 21027, Italy
- Department of Physics, University of Trieste, Trieste 34127, Italy
| | - Hendrik Vondracek
- Nano-Innovation Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Trieste 34149, Italy
| | - Nicola Cefarin
- Istituto Officina dei Materiali, Consiglio Nazionale delle Ricerche, CNR-IOM, Trieste 34149, Italy
| | - Giacomo Ceccone
- Institute for Health and Consumer Protection, European Commission - Joint Research Centre, Ispra 21027, Italy
| | - Luigi Calzolai
- Institute for Health and Consumer Protection, European Commission - Joint Research Centre, Ispra 21027, Italy
| | - Laura Ballerini
- Neuroscience Area, Scuola Internazionale Superiore di Studi Avanzati, Trieste 34136, Italy
| | - Pietro Parisse
- Nano-Innovation Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Trieste 34149, Italy
- Istituto Officina dei Materiali, Consiglio Nazionale delle Ricerche, CNR-IOM, Trieste 34149, Italy
| | - Loredana Casalis
- Nano-Innovation Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Trieste 34149, Italy
| |
Collapse
|
28
|
Andryszkiewicz W, Misiąg P, Karwowska A, Resler Z, Wojno A, Kulbacka J, Szewczyk A, Rembiałkowska N. Cancer Metastases to the Liver: Mechanisms of Tumor Cell Colonization. Pharmaceuticals (Basel) 2024; 17:1251. [PMID: 39338413 PMCID: PMC11434846 DOI: 10.3390/ph17091251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
The liver is one of the most common sites for metastasis, which involves the spread from primary tumors to surrounding organs and tissues in the human body. There are a few steps in cancer expansion: invasion, inflammatory processes allowing the hepatic niche to be created, adhesions to ECM, neovascularization, and secretion of enzymes. The spread of tumor cells depends on the microenvironment created by the contribution of many biomolecules, including proteolytic enzymes, cytokines, growth factors, and cell adhesion molecules that enable tumor cells to interact with the microenvironment. Moreover, the microenvironment plays a significant role in tumor growth and expansion. The secreted enzymes help cancer cells facilitate newly formed hepatic niches and promote migration and invasion. Our study discusses pharmacological methods used to prevent liver metastasis by targeting the tumor microenvironment and cancer cell colonization in the liver. We examine randomized studies focusing on median survival duration and median overall survival in patients administered placebo compared with those treated with bevacizumab, ramucirumab, regorafenib, and ziv-aflibercept in addition to current chemotherapy. We also include research on mice and their responses to these medications, which may suppress metastasis progression. Finally, we discuss the significance of non-pharmacological methods, including surgical procedures, radiotherapy, cryotherapy, radiofrequency ablation (RFA), and transarterial embolization (TAE). In conclusion, the given methods can successfully prevent metastases to the liver and prolong the median survival duration and median overall survival in patients suffering from cancer.
Collapse
Affiliation(s)
- Wiktoria Andryszkiewicz
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Piotr Misiąg
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Anna Karwowska
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Zofia Resler
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Aleksandra Wojno
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
29
|
Kabatnik S, Post F, Drici L, Bartels AS, Strauss MT, Zheng X, Madsen GI, Mund A, Rosenberger FA, Moreira J, Mann M. Spatial characterization and stratification of colorectal adenomas by deep visual proteomics. iScience 2024; 27:110620. [PMID: 39252972 PMCID: PMC11381895 DOI: 10.1016/j.isci.2024.110620] [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: 12/22/2023] [Revised: 05/13/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Colorectal adenomas (CRAs) are potential precursor lesions to adenocarcinomas, currently classified by morphological features. We aimed to establish a molecular feature-based risk allocation framework toward improved patient stratification. Deep visual proteomics (DVP) is an approach that combines image-based artificial intelligence with automated microdissection and ultra-high sensitive mass spectrometry. Here, we used DVP on formalin-fixed, paraffin-embedded (FFPE) CRA tissues from nine male patients, immunohistologically stained for caudal-type homeobox 2 (CDX2), a protein implicated in colorectal cancer, enabling the characterization of cellular heterogeneity within distinct tissue regions and across patients. DVP identified DMBT1, MARCKS, and CD99 as protein markers linked to recurrence, suggesting their potential for risk assessment. It also detected a metabolic shift to anaerobic glycolysis in cells with high CDX2 expression. Our findings underscore the potential of spatial proteomics to refine early stage detection and contribute to personalized patient management strategies and provided novel insights into metabolic reprogramming.
Collapse
Affiliation(s)
- Sonja Kabatnik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Frederik Post
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Lylia Drici
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Annette Snejbjerg Bartels
- Precision Cancer Medicine Laboratory, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maximilian T Strauss
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Xiang Zheng
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Gunvor I Madsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Andreas Mund
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Florian A Rosenberger
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - José Moreira
- Precision Cancer Medicine Laboratory, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
30
|
Kang Y, Wang Z, An K, Hou Q, Zhang Z, Su J. Introgression drives adaptation to the plateau environment in a subterranean rodent. BMC Biol 2024; 22:187. [PMID: 39218870 PMCID: PMC11368017 DOI: 10.1186/s12915-024-01986-y] [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: 01/27/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Introgression has repeatedly been shown to play an important role in the adaptation of species to extreme environments, yet how introgression enables rodents with specialized subterranean lifestyle to acclimatize to high altitudes is still unclear. Myospalacinae is a group of subterranean rodents, among which the high-altitude plateau zokors (Eospalax baileyi) and the low-altitude Gansu zokors (E. cansus) are sympatrically distributed in the grassland ecosystems of the Qinghai-Tibet Plateau (QTP). Together, they provide a model for the study of the role of introgression in the adaptation of low-altitude subterranean rodents to high altitudes. RESULTS Applying low-coverage whole-genome resequencing and population genetics analyses, we identified evidence of adaptive introgression from plateau zokors into Gansu zokors, which likely facilitated the adaptation of the latter to the high-altitude environment of the QTP. We identified positively selected genes with functions related to energy metabolism, cardiovascular system development, calcium ion transport, and response to hypoxia which likely made critical contributions to adaptation to the plateau environment in both plateau zokors and high-altitude populations of Gansu zokors. CONCLUSIONS Introgression of genes associated with hypoxia adaptation from plateau zokors may have played a role in the adaptation of Gansu zokors to the plateau environment. Our study provides new insights into the understanding of adaptive evolution of species on the QTP and the importance of introgression in the adaptation of species to high-altitude environments.
Collapse
Affiliation(s)
- Yukun Kang
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhicheng Wang
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Kang An
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Qiqi Hou
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhiming Zhang
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China
| | - Junhu Su
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, 730070, China.
- Gansu Agricultural University-Massey University Research Centre for Grassland Biodiversity, Gansu Agricultural University, Lanzhou, 730070, China.
- Gansu Qilianshan Grassland Ecosystem Observation and Research Station, Wuwei, 733200, China.
| |
Collapse
|
31
|
Zhang JY, Su YH, Wang X, Yao X, Du JZ. Recent Progress on Nanomedicine-Mediated Repolarization of Tumor-Associated Macrophages for Cancer Immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2001. [PMID: 39425549 DOI: 10.1002/wnan.2001] [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: 05/10/2024] [Revised: 08/07/2024] [Accepted: 09/18/2024] [Indexed: 10/21/2024]
Abstract
Tumor-associated macrophages (TAMs) constitute the largest number of immune cells in the tumor microenvironment (TME). They play an essential role in promoting tumor progression and metastasis, which makes them a potential therapeutic target for cancer treatment. TAMs are usually divided into two categories: pro-tumoral M2-like TAMs and antitumoral M1 phenotypes at either extreme. The reprogramming of M2-like TAMs toward a tumoricidal M1 phenotype is of particular interest for the restoration of antitumor immunity in cancer immunotherapy. Notably, nanomedicines have shown great potential for cancer therapy due to their unique structures and properties. This review will briefly describe the biological features and roles of TAMs in tumor, and then discuss recent advances in nanomedicine-mediated repolarization of TAMs for cancer immunotherapy. Finally, perspectives on nanomedicine-mediated repolarization of TAMs for effective cancer immunotherapy are also presented.
Collapse
Affiliation(s)
- Jing-Yang Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China
| | - Yun-He Su
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China
| | - Xu Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China
| | - Xueqing Yao
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
32
|
Kovale L, Singh MK, Kim J, Ha J. Role of Autophagy and AMPK in Cancer Stem Cells: Therapeutic Opportunities and Obstacles in Cancer. Int J Mol Sci 2024; 25:8647. [PMID: 39201332 PMCID: PMC11354724 DOI: 10.3390/ijms25168647] [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: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer stem cells represent a resilient subset within the tumor microenvironment capable of differentiation, regeneration, and resistance to chemotherapeutic agents, often using dormancy as a shield. Their unique properties, including drug resistance and metastatic potential, pose challenges for effective targeting. These cells exploit certain metabolic processes for their maintenance and survival. One of these processes is autophagy, which generally helps in energy homeostasis but when hijacked by CSCs can help maintain their stemness. Thus, it is often referred as an Achilles heel in CSCs, as certain cancers tend to depend on autophagy for survival. Autophagy, while crucial for maintaining stemness in cancer stem cells (CSCs), can also serve as a vulnerability in certain contexts, making it a complex target for therapy. Regulators of autophagy like AMPK (5' adenosine monophosphate-activated protein kinase) also play a crucial role in maintaining CSCs stemness by helping CSCs in metabolic reprogramming in harsh environments. The purpose of this review is to elucidate the interplay between autophagy and AMPK in CSCs, highlighting the challenges in targeting autophagy and discussing therapeutic strategies to overcome these limitations. This review focuses on previous research on autophagy and its regulators in cancer biology, particularly in CSCs, addresses the remaining unanswered questions, and potential targets for therapy are also brought to attention.
Collapse
Affiliation(s)
- Lochana Kovale
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Joungmok Kim
- Department of Oral Biochemistry and Molecular Biology, College of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| |
Collapse
|
33
|
Liu T, Yao W, Sun W, Yuan Y, Liu C, Liu X, Wang X, Jiang H. Components, Formulations, Deliveries, and Combinations of Tumor Vaccines. ACS NANO 2024; 18:18801-18833. [PMID: 38979917 DOI: 10.1021/acsnano.4c05065] [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: 07/10/2024]
Abstract
Tumor vaccines, an important part of immunotherapy, prevent cancer or kill existing tumor cells by activating or restoring the body's own immune system. Currently, various formulations of tumor vaccines have been developed, including cell vaccines, tumor cell membrane vaccines, tumor DNA vaccines, tumor mRNA vaccines, tumor polypeptide vaccines, virus-vectored tumor vaccines, and tumor-in-situ vaccines. There are also multiple delivery systems for tumor vaccines, such as liposomes, cell membrane vesicles, viruses, exosomes, and emulsions. In addition, to decrease the risk of tumor immune escape and immune tolerance that may exist with a single tumor vaccine, combination therapy of tumor vaccines with radiotherapy, chemotherapy, immune checkpoint inhibitors, cytokines, CAR-T therapy, or photoimmunotherapy is an effective strategy. Given the critical role of tumor vaccines in immunotherapy, here, we look back to the history of tumor vaccines, and we discuss the antigens, adjuvants, formulations, delivery systems, mechanisms, combination therapy, and future directions of tumor vaccines.
Collapse
Affiliation(s)
- Tengfei Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyan Yao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yihan Yuan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Chen Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
34
|
Zhu T, Alves SM, Adamo A, Wen X, Corn KC, Shostak A, Johnson S, Shaub ND, Martello SE, Hacker BC, D'Amore A, Bardhan R, Rafat M. Mammary tissue-derived extracellular matrix hydrogels reveal the role of irradiation in driving a pro-tumor and immunosuppressive microenvironment. Biomaterials 2024; 308:122531. [PMID: 38531198 PMCID: PMC11065579 DOI: 10.1016/j.biomaterials.2024.122531] [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: 12/18/2023] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Radiation therapy (RT) is essential for triple negative breast cancer (TNBC) treatment. However, patients with TNBC continue to experience recurrence after RT. The role of the extracellular matrix (ECM) of irradiated breast tissue in tumor recurrence is still unknown. In this study, we evaluated the structure, molecular composition, and mechanical properties of irradiated murine mammary fat pads (MFPs) and developed ECM hydrogels from decellularized tissues (dECM) to assess the effects of RT-induced ECM changes on breast cancer cell behavior. Irradiated MFPs were characterized by increased ECM deposition and fiber density compared to unirradiated controls, which may provide a platform for cell invasion and proliferation. ECM component changes in collagens I, IV, and VI, and fibronectin were observed following irradiation in both MFPs and dECM hydrogels. Encapsulated TNBC cell proliferation and invasive capacity was enhanced in irradiated dECM hydrogels. In addition, TNBC cells co-cultured with macrophages in irradiated dECM hydrogels induced M2 macrophage polarization and exhibited further increases in proliferation. Our study establishes that the ECM in radiation-damaged sites promotes TNBC invasion and proliferation as well as an immunosuppressive microenvironment. This work represents an important step toward elucidating how changes in the ECM after RT contribute to breast cancer recurrence.
Collapse
Affiliation(s)
- Tian Zhu
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Steven M Alves
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Arianna Adamo
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kevin C Corn
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Anastasia Shostak
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Nicholas D Shaub
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shannon E Martello
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Benjamin C Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Antonio D'Amore
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
35
|
Mastronikolis NS, Kyrodimos E, Piperigkou Z, Spyropoulou D, Delides A, Giotakis E, Alexopoulou M, Bakalis NA, Karamanos NK. Matrix-based molecular mechanisms, targeting and diagnostics in oral squamous cell carcinoma. IUBMB Life 2024; 76:368-382. [PMID: 38168122 DOI: 10.1002/iub.2803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a head and neck cancer (HNC) with a high mortality rate. OSCC is developed in the oral cavity and it is triggered by many etiologic factors and can metastasize both regionally and distantly. Recent research advances in OSCC improved our understanding on the molecular mechanisms involved in and the initiation of OSCC metastasis. The key roles of the extracellular matrix (ECM) in OSCC are an emerging area of intensive research as the ECM macromolecular network is actively involved in events that regulate cellular morphological and functional properties, transcription and cell signaling mechanisms in invasion and metastasis. The provisional matrix that is formed by cancer cells is profoundly different in composition and functions as compared with the matrix of normal tissue. Fibroblasts are mainly responsible for matrix production and remodeling, but in cancer, the tumor matrix in the tumor microenvironment (TME) also originates from cancer cells. Even though extensive research has been conducted on the role of ECM in regulating cancer pathogenesis, its role in modulating OSCC is less elucidated since there are several issues yet to be fully understood. This critical review is focused on recent research as to present and discuss on the involvement of ECM macromolecular effectors (i.e., proteoglycans, integrins, matrix metalloproteinases) in OSCC development and progression.
Collapse
Affiliation(s)
- Nicholas S Mastronikolis
- Department of Otorhinolaryngology - Head and Neck Surgery, School of Medicine, University of Patras, Patras, Greece
| | - Efthymios Kyrodimos
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology - Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Despoina Spyropoulou
- Department of Radiation Oncology, School of Medicine, University of Patras, Patras, Greece
| | - Alexander Delides
- 2nd Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Attikon' University Hospital, Athens, Greece
| | - Evangelos Giotakis
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
- Department of Radiation Oncology, School of Medicine, University of Patras, Patras, Greece
- 2nd Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Attikon' University Hospital, Athens, Greece
| | - Miranda Alexopoulou
- Department of Maxillofacial Surgery, University Hospital of Patras, Patras, Greece
| | - Nick A Bakalis
- Department of Nursing, University of Patras, Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology - Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
36
|
Thakur C, Qiu Y, Pawar A, Chen F. Epigenetic regulation of breast cancer metastasis. Cancer Metastasis Rev 2024; 43:597-619. [PMID: 37857941 DOI: 10.1007/s10555-023-10146-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Breast cancer is the most frequently diagnosed malignancy and the second leading cause of cancer-related mortality among women worldwide. Recurrent metastasis is associated with poor patient outcomes and poses a significant challenge in breast cancer therapies. Cancer cells adapting to a new tissue microenvironment is the key event in distant metastasis development, where the disseminating tumor cells are likely to acquire genetic and epigenetic alterations during the process of metastatic colonization. Despite several decades of research in this field, the exact mechanisms governing metastasis are not fully understood. However, emerging body of evidence indicates that in addition to genetic changes, epigenetic reprogramming of cancer cells and the metastatic niche are paramount toward successful metastasis. Here, we review and discuss the latest knowledge about the salient attributes of metastasis and epigenetic regulation in breast cancer and crucial research domains that need further investigation.
Collapse
Affiliation(s)
- Chitra Thakur
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA.
| | - Yiran Qiu
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA
| | - Aashna Pawar
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA
| | - Fei Chen
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA.
| |
Collapse
|
37
|
Conboy JP, Istúriz Petitjean I, van der Net A, Koenderink GH. How cytoskeletal crosstalk makes cells move: Bridging cell-free and cell studies. BIOPHYSICS REVIEWS 2024; 5:021307. [PMID: 38840976 PMCID: PMC11151447 DOI: 10.1063/5.0198119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cell migration is a fundamental process for life and is highly dependent on the dynamical and mechanical properties of the cytoskeleton. Intensive physical and biochemical crosstalk among actin, microtubules, and intermediate filaments ensures their coordination to facilitate and enable migration. In this review, we discuss the different mechanical aspects that govern cell migration and provide, for each mechanical aspect, a novel perspective by juxtaposing two complementary approaches to the biophysical study of cytoskeletal crosstalk: live-cell studies (often referred to as top-down studies) and cell-free studies (often referred to as bottom-up studies). We summarize the main findings from both experimental approaches, and we provide our perspective on bridging the two perspectives to address the open questions of how cytoskeletal crosstalk governs cell migration and makes cells move.
Collapse
Affiliation(s)
- James P. Conboy
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Irene Istúriz Petitjean
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Anouk van der Net
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Gijsje H. Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| |
Collapse
|
38
|
Fu J, Lin J, Dai Z, Lin B, Zhang J. Hypoxia-associated autophagy flux dysregulation in human cancers. Cancer Lett 2024; 590:216823. [PMID: 38521197 DOI: 10.1016/j.canlet.2024.216823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/09/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
A general feature of cancer is hypoxia, determined as low oxygen levels. Low oxygen levels may cause cells to alter in ways that contribute to tumor growth and resistance to treatment. Hypoxia leads to variations in cancer cell metabolism, angiogenesis and metastasis. Furthermore, a hypoxic tumor microenvironment might induce immunosuppression. Moreover, hypoxia has the potential to impact cellular processes, such as autophagy. Autophagy refers to the catabolic process by which damaged organelles and toxic macromolecules are broken down. The abnormal activation of autophagy has been extensively recorded in human tumors and it serves as a regulator of cell growth, spread to other parts of the body, and resistance to treatment. There is a correlation between hypoxia and autophagy in human malignancies. Hypoxia can regulate the activity of AMPK, mTOR, Beclin-1, and ATGs to govern autophagy in human malignancies. Furthermore, HIF-1α, serving as an indicator of low oxygen levels, controls the process of autophagy. Hypoxia-induced autophagy has a crucial role in regulating the growth, spread, and resistance to treatment in human malignancies. Hypoxia-induced regulation of autophagy can impact other mechanisms of cell death, such as apoptosis. Chemoresistance and radioresistance have become significant challenges in recent years. Hypoxia-mediated autophagy plays a crucial role in determining the response to these therapeutic treatments.
Collapse
Affiliation(s)
- Jiding Fu
- Department of Intensive Care Unit, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Zili Dai
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Baisheng Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jian Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China.
| |
Collapse
|
39
|
Zuo Q, Wu Y, Hu Y, Shao C, Liang Y, Chen L, Guo Q, Huang P, Chen Q. Targeting lipid reprogramming in the tumor microenvironment by traditional Chinese medicines as a potential cancer treatment. Heliyon 2024; 10:e30807. [PMID: 38765144 PMCID: PMC11101863 DOI: 10.1016/j.heliyon.2024.e30807] [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: 11/13/2023] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024] Open
Abstract
In the last ten years, there has been a notable rise in the study of metabolic abnormalities in cancer cells. However, compared to glucose or glutamine metabolism, less attention has been paid to the importance of lipid metabolism in tumorigenesis. Recent developments in lipidomics technologies have allowed for detailed analysis of lipid profiles within cancer cells and other cellular players present within the tumor microenvironment (TME). Traditional Chinese medicine (TCM) and its bioactive components have a long history of use in cancer treatments and are also being studied for their potential role in regulating metabolic reprogramming within TME. This review focuses on four core abnormalities altered by lipid reprogramming in cancer cells: de novo synthesis and exogenous uptake of fatty acids (FAs), upregulated fatty acid oxidation (FAO), cholesterol accumulation, which offer benefits for tumor growth and metastasis. The review also discusses how altered lipid metabolism impacts infiltrating immune cell function and phenotype as these interactions between cancer-stromal become more pronounced during tumor progression. Finally, recent literature is highlighted regarding how cancer cells can be metabolically reprogrammed by specific Chinese herbal components with potential therapeutic benefits related to lipid metabolic and signaling pathways.
Collapse
Affiliation(s)
- Qian Zuo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yingchao Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuyu Hu
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| | - Cui Shao
- The First Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqi Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liushan Chen
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| | - Qianqian Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Ping Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qianjun Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
40
|
Sun S, Liu Y, Gao H, Guan W, Zhao Y, Li G. Cell culture on suspended fiber for tissue regeneration: A review. Int J Biol Macromol 2024; 268:131827. [PMID: 38670204 DOI: 10.1016/j.ijbiomac.2024.131827] [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: 12/25/2023] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Cell culturing is a cornerstone of tissue engineering, playing a crucial role in tissue regeneration, drug screening, and the study of disease mechanisms. Among various culturing techniques, 3D culture systems, particularly those utilizing suspended fiber scaffolds, offer a more physiologically relevant environment than traditional 2D monolayer cultures. These 3D scaffolds enhance cell growth, differentiation, and proliferation by mimicking the in vivo cellular milieu. This review focuses on the critical role of suspended fiber scaffolds in tissue engineering. We compare the effectiveness of 3D suspended fiber scaffolds with 2D culture systems, discussing their respective benefits and limitations in the context of tissue regeneration. Furthermore, we explore the preparation methods of suspended fiber scaffolds and their potential applications. The review concludes by considering future research directions for optimizing suspended fiber scaffolds to address specific challenges in tissue regeneration, underscoring their significant promise in advancing tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Shaolan Sun
- Co-innovation Center of Neuroregeneration, Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, PR China
| | - Yaqiong Liu
- Co-innovation Center of Neuroregeneration, Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, PR China
| | - Hongxia Gao
- Co-innovation Center of Neuroregeneration, Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, PR China
| | - Wenchao Guan
- Co-innovation Center of Neuroregeneration, Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, PR China
| | - Yahong Zhao
- Co-innovation Center of Neuroregeneration, Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, PR China
| | - Guicai Li
- Co-innovation Center of Neuroregeneration, Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001 Nantong, PR China; NMPA Key Laboratory for Quality Evaluation of Medical Protective and Implant Devices, 450018 Zhengzhou, PR China.
| |
Collapse
|
41
|
Khosravi G, Mostafavi S, Bastan S, Ebrahimi N, Gharibvand RS, Eskandari N. Immunologic tumor microenvironment modulators for turning cold tumors hot. Cancer Commun (Lond) 2024; 44:521-553. [PMID: 38551889 PMCID: PMC11110955 DOI: 10.1002/cac2.12539] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 05/23/2024] Open
Abstract
Tumors can be classified into distinct immunophenotypes based on the presence and arrangement of cytotoxic immune cells within the tumor microenvironment (TME). Hot tumors, characterized by heightened immune activity and responsiveness to immune checkpoint inhibitors (ICIs), stand in stark contrast to cold tumors, which lack immune infiltration and remain resistant to therapy. To overcome immune evasion mechanisms employed by tumor cells, novel immunologic modulators have emerged, particularly ICIs targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1/programmed death-ligand 1(PD-1/PD-L1). These agents disrupt inhibitory signals and reactivate the immune system, transforming cold tumors into hot ones and promoting effective antitumor responses. However, challenges persist, including primary resistance to immunotherapy, autoimmune side effects, and tumor response heterogeneity. Addressing these challenges requires innovative strategies, deeper mechanistic insights, and a combination of immune interventions to enhance the effectiveness of immunotherapies. In the landscape of cancer medicine, where immune cold tumors represent a formidable hurdle, understanding the TME and harnessing its potential to reprogram the immune response is paramount. This review sheds light on current advancements and future directions in the quest for more effective and safer cancer treatment strategies, offering hope for patients with immune-resistant tumors.
Collapse
Affiliation(s)
- Gholam‐Reza Khosravi
- Department of Medical ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Samaneh Mostafavi
- Department of ImmunologyFaculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Sanaz Bastan
- Department of Medical ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Narges Ebrahimi
- Department of Medical ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Roya Safari Gharibvand
- Department of ImmunologySchool of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Nahid Eskandari
- Department of Medical ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
42
|
Tang H, Chen L, Liu X, Zeng S, Tan H, Chen G. Pan-cancer dissection of vasculogenic mimicry characteristic to provide potential therapeutic targets. Front Pharmacol 2024; 15:1346719. [PMID: 38694917 PMCID: PMC11061449 DOI: 10.3389/fphar.2024.1346719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/30/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction Vasculogenic mimicry (VM) represents a novel form of tumor angiogenesis that is associated with tumor invasiveness and drug resistance. However, the VM landscape across cancer types remains poorly understood. In this study, we elucidate the characterizations of VM across cancers based on multi-omics data and provide potential targeted therapeutic strategies. Methods Multi-omics data from The Cancer Genome Atlas was used to conduct comprehensive analyses of the characteristics of VM related genes (VRGs) across cancer types. Pan-cancer vasculogenic mimicry score was established to provide a depiction of the VM landscape across cancer types. The correlation between VM and cancer phenotypes was conducted to explore potential regulatory mechanisms of VM. We further systematically examined the relationship between VM and both tumor immunity and tumor microenvironment (TME). In addition, cell communication analysis based on single-cell transcriptome data was used to investigate the interactions between VM cells and TME. Finally, transcriptional and drug response data from the Genomics of Drug Sensitivity in Cancer database were utilized to identify potential therapeutic targets and drugs. The impact of VM on immunotherapy was also further clarified. Results Our study revealed that VRGs were dysregulated in tumor and regulated by multiple mechanisms. Then, VM level was found to be heterogeneous among different tumors and correlated with tumor invasiveness, metastatic potential, malignancy, and prognosis. VM was found to be strongly associated with epithelial-mesenchymal transition (EMT). Further analyses revealed cancer-associated fibroblasts can promote EMT and VM formation. Furthermore, the immune-suppressive state is associated with a microenvironment characterized by high levels of VM. VM score can be used as an indicator to predict the effect of immunotherapy. Finally, seven potential drugs targeting VM were identified. Conclusion In conclusion, we elucidate the characteristics and key regulatory mechanisms of VM across various cancer types, underscoring the pivotal role of CAFs in VM. VM was further found to be associated with the immunosuppressive TME. We also provide clues for the research of drugs targeting VM. Our study provides an initial overview and reference point for future research on VM, opening up new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Haibin Tang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liuxun Chen
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xvdong Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengjie Zeng
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Tan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
43
|
Major G, Ahn M, Cho WW, Santos M, Wise J, Phillips E, Wise SG, Jang J, Rnjak-Kovacina J, Woodfield T, Lim KS. Programming temporal stiffness cues within extracellular matrix hydrogels for modelling cancer niches. Mater Today Bio 2024; 25:101004. [PMID: 38420142 PMCID: PMC10900776 DOI: 10.1016/j.mtbio.2024.101004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Extracellular matrix (ECM) stiffening is a common occurrence during the progression of many diseases, such as breast cancer. To accurately mimic the pathophysiological context of disease within 3D in vitro models, there is high demand for smart biomaterials which replicate the dynamic and temporal mechanical cues of diseased states. This study describes a preclinical disease model, using breast cancer as an example, which replicates the dynamic plasticity of the tumour microenvironment by incorporating temporal (3-week progression) biomechanical cues within a tissue-specific hydrogel microenvironment. The composite hydrogel formulation, integrating adipose-derived decellularised ECM (AdECM) and silk fibroin, was initially crosslinked using a visible light-mediated system, and then progressively stiffened through spontaneous secondary structure interactions inherent between the polymer chains (∼10-15 kPa increase, with a final stiffness of 25 kPa). When encapsulated and cultured in vitro, MCF-7 breast cancer cells initially formed numerous, large spheroids (>1000 μm2 in area), however, with progressive temporal stiffening, cells demonstrated growth arrest and underwent phenotypic changes resulting in intratumoral heterogeneity. Unlike widely-investigated static mechanical models, this stiffening hydrogel allowed for progressive phenotypic changes to be observed, and fostered the development of mature organoid-like spheroids, which mimicked both the organisation and acinar-structures of mature breast epithelium. The spheroids contained a central population of cells which expressed aggressive cellular programs, evidenced by increased fibronectin expression and reduction of E-cadherin. The phenotypic heterogeneity observed using this model is more reflective of physiological tumours, demonstrating the importance of establishing temporal cues within preclinical models in future work. Overall, the developed model demonstrated a novel strategy to uncouple ECM biomechanical properties from the cellular complexities of the disease microenvironment and offers the potential for wide applicability in other 3D in vitro disease models through addition of tissue-specific dECM materials.
Collapse
Affiliation(s)
- Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Minjun Ahn
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Won-Woo Cho
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Miguel Santos
- Applied Materials Group, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jessika Wise
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Steven G Wise
- Applied Materials Group, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jinah Jang
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
- Tyree Institute of Health Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
- Light-Activated Materials Group, School of Medical Sciences, University of Sydney, Australia
| |
Collapse
|
44
|
Imodoye SO, Adedokun KA, Bello IO. From complexity to clarity: unravelling tumor heterogeneity through the lens of tumor microenvironment for innovative cancer therapy. Histochem Cell Biol 2024; 161:299-323. [PMID: 38189822 DOI: 10.1007/s00418-023-02258-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 01/09/2024]
Abstract
Despite the tremendous clinical successes recorded in the landscape of cancer therapy, tumor heterogeneity remains a formidable challenge to successful cancer treatment. In recent years, the emergence of high-throughput technologies has advanced our understanding of the variables influencing tumor heterogeneity beyond intrinsic tumor characteristics. Emerging knowledge shows that drivers of tumor heterogeneity are not only intrinsic to cancer cells but can also emanate from their microenvironment, which significantly favors tumor progression and impairs therapeutic response. Although much has been explored to understand the fundamentals of the influence of innate tumor factors on cancer diversity, the roles of the tumor microenvironment (TME) are often undervalued. It is therefore imperative that a clear understanding of the interactions between the TME and other tumor intrinsic factors underlying the plastic molecular behaviors of cancers be identified to develop patient-specific treatment strategies. This review highlights the roles of the TME as an emerging factor in tumor heterogeneity. More particularly, we discuss the role of the TME in the context of tumor heterogeneity and explore the cutting-edge diagnostic and therapeutic approaches that could be used to resolve this recurring clinical conundrum. We conclude by speculating on exciting research questions that can advance our understanding of tumor heterogeneity with the goal of developing customized therapeutic solutions.
Collapse
Affiliation(s)
- Sikiru O Imodoye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| | - Kamoru A Adedokun
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Ibrahim O Bello
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, King Saud University, Riyadh, Saudi Arabia.
- Department of Pathology, University of Helsinki, Haartmaninkatu 3, 00014, Helsinki, Finland.
| |
Collapse
|
45
|
Uddin MH, Zhang D, Muqbil I, El-Rayes BF, Chen H, Philip PA, Azmi AS. Deciphering cellular plasticity in pancreatic cancer for effective treatments. Cancer Metastasis Rev 2024; 43:393-408. [PMID: 38194153 DOI: 10.1007/s10555-023-10164-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
Cellular plasticity and therapy resistance are critical features of pancreatic cancer, a highly aggressive and fatal disease. The pancreas, a vital organ that produces digestive enzymes and hormones, is often affected by two main types of cancer: the pre-dominant ductal adenocarcinoma and the less common neuroendocrine tumors. These cancers are difficult to treat due to their complex biology characterized by cellular plasticity leading to therapy resistance. Cellular plasticity refers to the capability of cancer cells to change and adapt to different microenvironments within the body which includes acinar-ductal metaplasia, epithelial to mesenchymal/epigenetic/metabolic plasticity, as well as stemness. This plasticity allows heterogeneity of cancer cells, metastasis, and evasion of host's immune system and develops resistance to radiation, chemotherapy, and targeted therapy. To overcome this resistance, extensive research is ongoing exploring the intrinsic and extrinsic factors through cellular reprogramming, chemosensitization, targeting metabolic, key survival pathways, etc. In this review, we discussed the mechanisms of cellular plasticity involving cellular adaptation and tumor microenvironment and provided a comprehensive understanding of its role in therapy resistance and ways to overcome it.
Collapse
Affiliation(s)
- Md Hafiz Uddin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 740, Detroit, MI, 48201, USA.
| | - Dingqiang Zhang
- Department of Natural Sciences, Lawrence Technological University, 21000 W 10 Mile Rd, Southfield, MI, 48075, USA
| | - Irfana Muqbil
- Department of Natural Sciences, Lawrence Technological University, 21000 W 10 Mile Rd, Southfield, MI, 48075, USA
| | - Bassel F El-Rayes
- Division of Hematology and Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 740, Detroit, MI, 48201, USA
- Henry Ford Health Systems, Detroit, MI, 48202, USA
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 740, Detroit, MI, 48201, USA.
| |
Collapse
|
46
|
Miron RJ, Bohner M, Zhang Y, Bosshardt DD. Osteoinduction and osteoimmunology: Emerging concepts. Periodontol 2000 2024; 94:9-26. [PMID: 37658591 DOI: 10.1111/prd.12519] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/23/2023] [Accepted: 07/20/2023] [Indexed: 09/03/2023]
Abstract
The recognition and importance of immune cells during bone regeneration, including around bone biomaterials, has led to the development of an entire field termed "osteoimmunology," which focuses on the connection and interplay between the skeletal system and immune cells. Most studies have focused on the "osteogenic" capacity of various types of bone biomaterials, and much less focus has been placed on immune cells despite being the first cell type in contact with implantable devices. Thus, the amount of literature generated to date on this topic makes it challenging to extract needed information. This review article serves as a guide highlighting advancements made in the field of osteoimmunology emphasizing the role of the osteoimmunomodulatory properties of biomaterials and their impact on osteoinduction. First, the various immune cell types involved in bone biomaterial integration are discussed, including the prominent role of osteal macrophages (OsteoMacs) during bone regeneration. Thereafter, key biomaterial properties, including topography, wettability, surface charge, and adsorption of cytokines, growth factors, ions, and other bioactive molecules, are discussed in terms of their impact on immune responses. These findings highlight and recognize the importance of the immune system and osteoimmunology, leading to a shift in the traditional models used to understand and evaluate biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | | | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| | | |
Collapse
|
47
|
Riaz F, Zhang J, Pan F. Forces at play: exploring factors affecting the cancer metastasis. Front Immunol 2024; 15:1274474. [PMID: 38361941 PMCID: PMC10867181 DOI: 10.3389/fimmu.2024.1274474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Metastatic disease, a leading and lethal indication of deaths associated with tumors, results from the dissemination of metastatic tumor cells from the site of primary origin to a distant organ. Dispersion of metastatic cells during the development of tumors at distant organs leads to failure to comply with conventional treatments, ultimately instigating abrupt tissue homeostasis and organ failure. Increasing evidence indicates that the tumor microenvironment (TME) is a crucial factor in cancer progression and the process of metastatic tumor development at secondary sites. TME comprises several factors contributing to the initiation and progression of the metastatic cascade. Among these, various cell types in TME, such as mesenchymal stem cells (MSCs), lymphatic endothelial cells (LECs), cancer-associated fibroblasts (CAFs), myeloid-derived suppressor cells (MDSCs), T cells, and tumor-associated macrophages (TAMs), are significant players participating in cancer metastasis. Besides, various other factors, such as extracellular matrix (ECM), gut microbiota, circadian rhythm, and hypoxia, also shape the TME and impact the metastatic cascade. A thorough understanding of the functions of TME components in tumor progression and metastasis is necessary to discover new therapeutic strategies targeting the metastatic tumor cells and TME. Therefore, we reviewed these pivotal TME components and highlighted the background knowledge on how these cell types and disrupted components of TME influence the metastatic cascade and establish the premetastatic niche. This review will help researchers identify these altered components' molecular patterns and design an optimized, targeted therapy to treat solid tumors and restrict metastatic cascade.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Jing Zhang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
48
|
Bhattacharya B, Nag S, Mukherjee S, Kulkarni M, Chandane P, Mandal D, Mukerjee N, Mirgh D, Anand K, Adhikari MD, Gorai S, Thorat N. Role of Exosomes in Epithelial-Mesenchymal Transition. ACS APPLIED BIO MATERIALS 2024; 7:44-58. [PMID: 38108852 PMCID: PMC10792609 DOI: 10.1021/acsabm.3c00941] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/19/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a fundamental process driving cancer metastasis, transforming non-motile cells into a motile population that migrates to distant organs and forms secondary tumors. In recent years, cancer research has revealed a strong connection between exosomes and the EMT. Exosomes, a subpopulation of extracellular vesicles, facilitate cellular communication and dynamically regulate various aspects of cancer metastasis, including immune cell suppression, extracellular matrix remodeling, metastasis initiation, EMT initiation, and organ-specific metastasis. Tumor-derived exosomes (TEXs) and their molecular cargo, comprising proteins, lipids, nucleic acids, and carbohydrates, are essential components that promote EMT in cancer. TEXs miRNAs play a crucial role in reprogramming the tumor microenvironment, while TEX surface integrins contribute to organ-specific metastasis. Exosome-based cancer metastasis research offers a deeper understanding about cancer and an effective theranostic platform development. Additionally, various therapeutic sources of exosomes are paving the way for innovative cancer treatment development. In this Review, we spotlight the role of exosomes in EMT and their theranostic impact, aiming to inspire cancer researchers worldwide to explore this fascinating field in more innovative ways.
Collapse
Affiliation(s)
- Bikramjit Bhattacharya
- Department
of Applied Microbiology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Sagnik Nag
- Department
of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore, Tamil Nadu 632014, India
| | - Sayantanee Mukherjee
- Amrita
School of NanoSciences and Molecular Medicine, Amrita Institute of Medical Sciences, Kochi, Kerala 682041, India
| | - Mrunal Kulkarni
- Department
of Pharmacy, BITS Pilani, Pilani, Rajasthan 333031, India
| | - Priti Chandane
- Department
of Biochemistry, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Debashmita Mandal
- Department
of Biotechnology, Maulana Abul Kalam Azad
University of Technology (MAKAUT), Haringhata, Nadia, West Bengal 741249, India
| | - Nobendu Mukerjee
- Center
for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
- Department
of Health Sciences, Novel Global Community
and Educational Foundation, Hebersham, New South Wales 2770, Australia
| | - Divya Mirgh
- Vaccine
and Immunotherapy Canter, Massachusetts
General Hospital, Boston, Massachusetts 02114, United States
| | - Krishnan Anand
- Department
of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Manab Deb Adhikari
- Department
of Biotechnology, University of North Bengal
Raja Rammohunpur, Darjeeling, West Bengal 734013, India
| | - Sukhamoy Gorai
- Rush University Medical
Center, 1620 W. Harrison St., Chicago, Illinois 60612, United States
| | - Nanasaheb Thorat
- Limerick
Digital Cancer Research Centre and Department of Physics, Bernal Institute, University of Limerick, Limerick V94T9PX, Ireland
| |
Collapse
|
49
|
Wang C, Ji J, Jin Y, Sun Y, Cai Q, Jiang J, Guo L, Zhou C, Zhang J. Tumor-mesothelium HOXA11-PDGF BB/TGF β1-miR-181a-5p-Egr1 feedforward amplifier circuity propels mesothelial fibrosis and peritoneal metastasis of gastric cancer. Oncogene 2024; 43:171-188. [PMID: 37989866 PMCID: PMC10786717 DOI: 10.1038/s41388-023-02891-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
A proportion of gastric cancer (GC) patients suffer from peritoneal metastasis (PM) in the late stage of tumor and these patients have a poor prognosis. To provide more care for GC patient with PM, a deeper exploration of the molecular characteristics of GC-PM is needed. Here we performed the in vitro and in vivo study to illustrate the effect of HOXA11 over-expressed GC cells on peritoneal mesothelial cells (HMrSV5), transcriptomics analyses of HMrSV5 cells co-cultured with HOXA11 over-expressed GC cells, counterparts or alone, cytokine array analyses of serum-free culture medium of HOXA11 over-expressed GC cells, we validated our findings through genetic manipulation of HMrSV5 cells and neutralizing antibodies targeting cytokines secreted by HOXA11 over-expressed GC cells in vitro, as well as utilized human peritoneal metastatic lesions to validate expression of potential targets. We identified that HOXA11 over-expressed GC cells strongly propelled mesothelial fibrosis in vivo and in vitro, and HOXA11 regulated paracrine and autocrine of PDGF BB and TGF β1 in GC cells to propel mesothelial fibrosis. Meanwhile, HOXA11 over-expressed GC cells drove PDGF BB and TGF β1 secretion to activate developmental-process related genes in HMrSV5 cells, including Egr1, which processes dependent on miR-181a-5p. Then, Egr1 could mediate peritoneal mesothelial fibrosis. Correspondingly, Egr1 over-expressed HMrSV5 cells supported migration and peritoneal dissemination of GC cells. Together our results suggest that a feedforward amplifier circuity governing GC cells and mesothelial cells in peritoneum contribute to peritoneal metastasis of GC cells.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Jun Ji
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Yangbing Jin
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Ying Sun
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Qu Cai
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Jinling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Liting Guo
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Chenfei Zhou
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| |
Collapse
|
50
|
Wang Y, Huang H, Weng H, Jia C, Liao B, Long Y, Yu F, Nie Y. Talin mechanotransduction in disease. Int J Biochem Cell Biol 2024; 166:106490. [PMID: 37914021 DOI: 10.1016/j.biocel.2023.106490] [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: 07/29/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
Talin protein (Talin 1/2) is a mechanosensitive cytoskeleton protein. The unique structure of the Talin plays a vital role in transmitting mechanical forces. Talin proteins connect the extracellular matrix to the cytoskeleton by linking to integrins and actin, thereby mediating the conversion of mechanical signals into biochemical signals and influencing disease progression as potential diagnostic indicators, therapeutic targets, and prognostic indicators of various diseases. Most studies in recent years have confirmed that mechanical forces also have a crucial role in the development of disease, and Talin has been found to play a role in several diseases. Still, more studies need to be done on how Talin is involved in mechanical signaling in disease. This review focuses on the mechanical signaling of Talin in disease, aiming to summarize the mechanisms by which Talin plays a role in disease and to provide references for further studies.
Collapse
Affiliation(s)
- Yingzi Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Haozhong Huang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Huimin Weng
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Chunsen Jia
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China; Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yongmei Nie
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China.
| |
Collapse
|