1
|
Lu Z, Lyu Z, Dong P, Liu Y, Huang L. N6-methyladenosine RNA modification in stomach carcinoma: Novel insights into mechanisms and implications for diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167793. [PMID: 40088577 DOI: 10.1016/j.bbadis.2025.167793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
N6-methyladenosine (m6A) RNA methylation is crucially involved in the genesis and advancement of gastric cancer (GC) by controlling various pathobiological aspects including gene expression, signal transduction, metabolism, cell death, epithelial-mesenchymal transition, angiogenesis, and exosome function. Despite its importance, the exact mechanisms by which m6A modification influences GC biology remain inadequately explored. This review consolidates the latest advances in uncovering the mechanisms and diverse roles of m6A in GC and proposes new research and translational directions. Key regulators (writers, readers, and erasers) of m6A, such as METTL3/14/16 and WTAP, significantly affect cancer progression, anticancer immune response, and treatment outcomes. m6A modification also impacts immune cell infiltration and the tumor microenvironment, highlighting its potential as a diagnostic and prognostic marker. Interactions between m6A methylation and non-coding RNAs offer further novel insights into GC development and therapeutic targets. Targeting m6A regulators could enhance immunotherapy response, overcome treatment resistance, and improve oncological and clinical outcomes. Models based on m6A can precisely predict treatment response and prognosis in GC. Additional investigation is needed to fully understand the mechanisms of m6A methylation and its potential clinical applications and relevance (e.g., as precise markers for early detection, prediction of outcome, and response to therapy and as therapeutic targets) in GC. Future research should focus on in vivo studies, potential clinical trials, and the examination of m6A modification in other types of cancers.
Collapse
Affiliation(s)
- Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaojie Lyu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Shanghai Institute of Pancreatic Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
2
|
Li Z, Wang T, Du S, Miao Z, Zhao Y, Tang Y, Meng X, Yu S, Zhang D, Jiang H, Du K, Wei W, Deng H. Tgm2-Catalyzed Covalent Cross-Linking of IκBα Drives NF-κB Nuclear Translocation to Promote SASP in Senescent Microglia. Aging Cell 2025; 24:e14463. [PMID: 39749582 PMCID: PMC12073898 DOI: 10.1111/acel.14463] [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: 07/30/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 01/04/2025] Open
Abstract
Microglia, as resident immune cells in the central nervous system (CNS), play a crucial role in maintaining homeostasis and phagocytosing metabolic waste in the brain. Senescent microglia exhibit decreased phagocytic capacity and increased neuroinflammation through senescence-associated secretory phenotype (SASP). This process contributes to the development of various neurodegenerative diseases, including Alzheimer's disease (AD). In this study, we found that SASP was elevated in senescent microglia, and proteomics showed that Tgm2 was upregulated. Mechanistically, we revealed that Tgm2-catalyzed covalent cross-linking of IκBα at K22 and Q248 residues in the cytoplasm of microglia, resulting in the reduction of IκBα and nuclear translocation of NF-κB to promote SASP production. Treatment of senescent microglia with Tgm2 inhibitors (Tg2-IN1 and Cys-D) resulted in reduced NF-κB nuclear translocation and decreased SASP. Additionally, oral administration of Cys-D significantly improved the aging phenotype in aged mice. To summarize, Tgm2 is a potential target for antiaging, and inhibitors of Tgm2 can serve as novel prophylactics or senomorphics.
Collapse
Affiliation(s)
- Zhiqiang Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Tianxiang Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Sijing Du
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Zelong Miao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Yujiao Zhao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Yuxiang Tang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Shangcheng Yu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Dongyuan Zhang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Hao Jiang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Kunlin Du
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| | - Wei Wei
- Wangjing Hospital, China Academy of Chinese Medical SciencesBeijingPeople's Republic of China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life SciencesTsinghua UniversityBeijingPeople's Republic of China
| |
Collapse
|
3
|
Yang Y, Kang Z, Cai J, Jia S, Fan S, Zhu H. Role of FHOD1 in tumor cells and tumor immune microenvironment. Front Immunol 2025; 16:1514488. [PMID: 40364836 PMCID: PMC12069282 DOI: 10.3389/fimmu.2025.1514488] [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: 10/21/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
FHOD 1 (Formin homology 2 domain containing protein 1) is a member of Diaphanous-related formins (DRFs) which contains a GTP-binding domain (GBD), formin homology (FH) 1 and FH 2 domains, a coiled-coil, and a diaphanous-like autoregulatory domain. Studies have shown that FHOD1 can not only regulate intracellular signals in tumor cells but also regulate various components of the tumor microenvironment (TME), such as T cells, B cells, cancer-associated fibroblasts (CAFs), some cytokines. Aberrant expression and dysfunction of the FHOD1 protein play a key role in tumor immunosuppression. Specifically, FHOD1 can impair function of chemokine receptors that are supposed to direct immune cells to localize to the tumor site accurately. As a result of this impairment, immune cells cannot migrate efficiently into TME, thereby impairing their ability to attack tumor cells. In addition, FHOD1 activated signaling pathways within the immune cells abnormally, resulting in their inability to recognize and destroy tumor cells effectively. Therefore, FHOD1 ultimately leads to a state of immunosuppression in TME, providing favorable conditions for the growth and spread of tumor cells. Altogether this review provides an in-depth understanding of the role of FHOD1 in tumor immunosuppression.
Collapse
Affiliation(s)
| | | | | | | | | | - Huifang Zhu
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
4
|
Wu YY, Yu LSY, Zhou HY, Xue JC. Effect of HepG2-Derived Exosome with PDGF-D Knockdown on Transformation of Normal Fibroblasts into Tumor-Associated Fibroblasts in Liver Cancer. FRONT BIOSCI-LANDMRK 2025; 30:26045. [PMID: 40152369 DOI: 10.31083/fbl26045] [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: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND It is known that the transformation of liver cancer-mediated fibroblasts into cancer-related fibroblasts (CAFs) is beneficial to the development of liver cancer. However, the specific mechanism is still unclear. METHODS Human hepatocarcinoma (HepG2) cells were treated with short hairpin RNA (shRNA) of platelet-derived growth factor-D (shPDGF-D) vector, and the exosomes secreted by the cells were separated using ultracentrifugation and identified by using nanoparticle tracking analysis, transmission electron microscope, and western blot analysis. Exosomes were co-cultured with mouse primary fibroblasts, and then the activity, proliferation, cell cycle, migration, epithelial-mesenchymal transition- (EMT-) and CAF marker-related protein expression levels of fibroblasts were determined by cell counting kit-8 (CCK-8), immunofluorescence, flow cytometry, wound healing, real-time reverse transcription-PCR, and western blotting assays, respectively. Co-cultured fibroblasts were mixed with HepG2 cells and injected subcutaneously into mice to construct animal models. The size and weight of xenograft tumor and the expression of epithelial-mesenchymal transition- (EMT-), angiogenesis- and CAFs marker-related proteins were detected. RESULTS The exosomes inhibited the proliferation, migration, EMT, and induced cell cycle arrest, as well as decreased the expression of α-SMA, FAP, MMP-9, and VEGF in fibroblasts. In vivo, sh-PDGF-D inhibited tumor growth, reduced the expressions of CD31, vimentin, α-SMA, FAP, MMP9, and VEGF, and promoted the expression of E-cadherin. CONCLUSIONS Exosomes derived from HepG2 cells transfected with shPDGF-D prevent normal fibroblasts from transforming into CAFs, thus inhibiting angiogenesis and EMT of liver cancer.
Collapse
Affiliation(s)
- Yan-Yan Wu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Liu-Shen-Yan Yu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Han-Yu Zhou
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Jun-Chao Xue
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Luo D, Liu Y, Lu Z, Huang L. Targeted therapy and immunotherapy for gastric cancer: rational strategies, novel advancements, challenges, and future perspectives. Mol Med 2025; 31:52. [PMID: 39923010 PMCID: PMC11806620 DOI: 10.1186/s10020-025-01075-y] [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: 07/16/2024] [Accepted: 01/10/2025] [Indexed: 02/10/2025] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors worldwide, and its treatment has been a focus of medical research. Herein we systematically review the current status of and advancements in targeted therapy and immunotherapy for GC, which have emerged as important treatment strategies in recent years with great potential, and summarize the efficacy and safety of such treatments. Targeted therapies against key targets in GC, including epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR), have shown remarkable therapeutic efficacies by inhibiting tumor progression and/or blood supply. In particular, markable breakthroughs have been made in HER2-targeting drugs for HER2-positive GC patients. To address intrinsic and acquired resistances to HER2-targeting drugs, novel therapeutic agents including bispecific antibodies and antibody-drug conjugates (ADC) targeting HER2 have been developed. Immunotherapy enhances the recognition and elimination of cancer cells by activating body anticancer immune system. Programmed cell death protein 1 (PD-1) and programmed cell death-ligand 1 (PD-L1) antibodies are the most commonly used immunotherapeutic agents and have been used with some success in GC treatment. Innovative immunotherapy modalities, including adoptive immune cell therapy, tumor vaccines, and non-specific immunomodulators therapy, and oncolytic viruses have shown promise in early-stage clinical trials for GC. Clinical trials have supported that targeted therapy and immunotherapy can significantly improve the survival and quality of life of GC patients. However, the effects of such therapies need to be further improved and more personalized, with advancement in researches on tumor immune microenvironment. Further studies remain needed to address the issues of drug resistance and adverse events pertaining to such therapies for GC. The combined application of such therapies and individualized treatment strategies should be further explored with novel drugs developed, to provide more effective treatments for GC patients.
Collapse
Affiliation(s)
- Dong Luo
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Center of Structural Heart Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, 200444, China.
| | - Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
- National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
6
|
Wei G, Nie Y, Sun M, Zhou W, Zhao H, Chen F, Zhu C. Cancer-associated fibroblasts induce almonertinib resistance in non-small cell lung cancer. J Transl Med 2025; 23:42. [PMID: 39794783 PMCID: PMC11724582 DOI: 10.1186/s12967-024-06064-z] [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: 08/14/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Almonertinib is the initial third-generation EGFR-TKI in China, but its resistance mechanism is unknown. Cancer-associated fibroblasts (CAFs) are essential matrix components in the tumor microenvironment, but their impact on almonertinib resistance is unknown. This study aimed to explore the correlation between CAFs and almonertinib resistance in non-small cell lung cancer (NSCLC). METHODS The anti-cancer effects of almonertinib on NSCLC cells, as well as the reversal of these effects mediated by CAFs, were validated through phenotypic experiments. Differential gene expression analysis, along with GO and KEGG enrichment analyses, was performed to predict the potential mechanisms underlying resistance to third-generation EGFR-TKIs. Finally, qPCR and Western blot analyses were used to explore the signaling pathways by which CAFs induce resistance to almonertinib in NSCLC cells. RESULTS Our findings revealed that almonertinib significantly suppressed the invasion, migration, and proliferation of EGFR T790M-mutant NSCLC cells. TGF-β1 successfully induced the differentiation of CAFs and upregulated the expression of CAF markers, including α-SMA and fibroblast activation protein (FAP). Exposure of H1975 cells to almonertinib increased TGF-β1 secretion. Additionally, CAFs enhanced the survival of almonertinib-treated NSCLC cells, whereas normal fibroblasts (NFs) exerted the opposite effect. qPCR analysis demonstrated that the expression of the core molecules of the Hippo pathway, YAP and TAZ, was lower in A549 cells than in H1975 cells, and CAF intervention further reduced YAP/TAZ expression in H1975 cells. Western blot analysis confirmed a significant reduction in YAP/TAZ protein levels in cancer cells treated with CAF-conditioned medium (CAF-CM) compared to those treated with normal control-conditioned medium (NC-CM). Finally, we demonstrated that CAFs induced resistance to almonertinib in NSCLC cells, potentially through a mechanism involving YAP/TAZ. CONCLUSION This study demonstrated that H1975 cells stimulated by almonertinib promoted the accumulation of CAFs in NSCLC cells, likely through increased secretion of TGF-β1. The accumulation of CAFs enhanced the survival of NSCLC cells undergoing almonertinib treatment and induced drug resistance. Additionally, the mechanism underlying CAF-induced drug resistance in NSCLC cells was potentially linked to the activation of the YAP/TAZ signaling pathway.
Collapse
Affiliation(s)
- Guohao Wei
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Yu Nie
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Min Sun
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Wenzheng Zhou
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Huihui Zhao
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China.
- Department of Oncology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| | - Fangfang Chen
- Department of Clinical Laboratory, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Chuandong Zhu
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| |
Collapse
|
7
|
Zhang X, Zhang M, Sun H, Wang X, Wang X, Sheng W, Xu M. The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells. J Adv Res 2025; 67:121-132. [PMID: 38309692 PMCID: PMC11725164 DOI: 10.1016/j.jare.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Transcription factors (TFs) fulfill a critical role in the formation and maintenance of different cell types during the developmental process as well as disease. It is believed that cancer-associated fibroblasts (CAFs) are activation status of tissue-resident fibroblasts or derived from form other cell types via transdifferentiation or dedifferentiation. Despite a subgroup of CAFs exhibit anti-cancer effects, most of them are reported to exert effects on tumor progression, further indicating their heterogeneous origin. AIM OF REVIEW This review aimed to summarize and review the roles of TFs in the reciprocal crosstalk between CAFs and tumor cells, discuss the emerging mechanisms, and their roles in cell-fate decision, cellular reprogramming and advancing our understanding of the gene regulatory networks over the period of cancer initiation and progression. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript delves into the key contributory factors of TFs that are involved in activating CAFs and maintaining their unique states. Additionally, it explores how TFs play a pivotal and multifaceted role in the reciprocal crosstalk between CAFs and tumor cells. This includes their involvement in processes such as epithelial-mesenchymal transition (EMT), proliferation, invasion, and metastasis, as well as metabolic reprogramming. TFs also have a role in constructing an immunosuppressive microenvironment, inducing resistance to radiation and chemotherapy, facilitating angiogenesis, and even 'educating' CAFs to support the malignancies of tumor cells. Furthermore, this manuscript delves into the current status of TF-targeted therapy and considers the future directions of TFs in conjunction with anti-CAFs therapies to address the challenges in clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Qi Y, Li R, Han M. Tumor-associated macrophages induce epithelial-mesenchymal transition and promote lung metastasis in breast cancer by activating the IL-6/STAT3/TGM2 axis. Int Immunopharmacol 2024; 143:113387. [PMID: 39426226 DOI: 10.1016/j.intimp.2024.113387] [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: 07/18/2024] [Revised: 09/28/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Breast cancer is one of the most common tumors in the world and metastasis is the major cause of tumor-related death. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and often associated with cancer metastasis. Nevertheless, the mechanism by which TAMs regulate breast cancer metastasis remain unclear. In this study, we found that transglutaminase 2 (TGM2) could serve as a crucial target in the modulation of TAMs-induced epithelial-mesenchymal transition (EMT) and invasion of breast cancer cells. Further analysis revealed that IL-6 secreted from TAMs, which was capable of inducing TGM2 expression through the activation of the JAK/STAT3 signaling pathway. Subsequent luciferase reporter assays demonstrated that STAT3 binds to the TGM2 promoter region, thereby transcriptionally enhancing TGM2 expression. In conclusion, our current research has identified the IL-6/STAT3/TGM2 axis as a pivotal regulator in breast tumorigenesis caused by TAMs, presenting a novel target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yana Qi
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| | - Ranran Li
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Mingyong Han
- Cancer therapy and Research Center, Shandong Provincal Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
9
|
Liu B, Cai Z, Wang Y, Liu X, Zhang B, Zheng Q, Li J, Li C, Cui Y, Lv P, Yang D. Transglutaminase 2 regulates endothelial cell calcification via IL-6-mediated autophagy. Front Pharmacol 2024; 15:1393534. [PMID: 39654623 PMCID: PMC11625581 DOI: 10.3389/fphar.2024.1393534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/31/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Endothelial cell (EC) calcification is an important marker of atherosclerotic calcification. ECs play a critical role not only in atherogenesis but also in intimal calcification, as they have been postulated to serve as a source of osteoprogenitor cells that initiate this process. While the role of transglutaminase 2 (TG2) in cellular differentiation, survival, apoptosis, autophagy, and cell adhesion is well established, the mechanism underlying the TG2-mediated regulation of EC calcification is yet to be fully elucidated. Methods The TG2 gene was overexpressed or silenced by using siRNA and recombinant adenovirus. RT-PCR and WB were used to analyze the relative expression of target genes and proteins. 5-BP method analyzed TG2 activity. mCherry-eGFP-LC3 adenovirus and transmission electron microscopy analyzed EC autophagy level. Calcium concentrations were measured by using a calcium colorimetric assay kit. Alizarin red S staining assay analyzed EC calcification level. Elisa analyzed IL-6 level. Establishing EC calcification model by using a calcification medium (CM). Results Our findings demonstrated that CM increased TG2 activity and expression, which activated the NF-κB signaling pathway, and induced IL-6 autocrine signaling in ECs. Furthermore, IL-6 activated the JAK2/STAT3 signaling pathway to suppress cell autophagy and promoted ECs calcification. Discussion ECs are not only critical for atherogenesis but also believed to be a source of osteoprogenitor cells that initiate intimal calcification. Previous research has shown that TG2 plays an important role in the development of VC, but the mechanism by which it exerts this effect is not yet fully understood. Our results demonstrated that TG2 forms complexes with NF-κB components inhibition of autophagy promoted endothelial cell calcification through EndMT. Therefore, our research investigated the molecular mechanism of EC calcification, which can provide new insights into the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Bo Liu
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiyuan Cai
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Wang
- The First Department of Ocular Fundus Diseases, Zhengzhou Second Hospital, Zhengzhou, Henan, China
| | - Xinye Liu
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Zhang
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Zheng
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jingye Li
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Cien Li
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanbo Cui
- Translational Medical Center, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Pengju Lv
- Department of clinical laboratory, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Dongwei Yang
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Lin W, Tang L, Zhuo C, Mao X, Shen J, Huang S, Li S, Qin Y, Liao J, Chen Y, Zhang X, Li Y, Song J, Meng L, Dong X, Li Y. scRNA-Seq Analysis Revealed CAFs Regulating HCC Cells via PTN Signaling. J Hepatocell Carcinoma 2024; 11:2269-2281. [PMID: 39582814 PMCID: PMC11583788 DOI: 10.2147/jhc.s493675] [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: 09/10/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) play a pivotal role in shaping the microenvironment of hepatocellular carcinoma (HCC). However, the mechanisms through which CAFs influence the progression of HCC remain incompletely understood. Methods Single-cell RNA sequencing datasets (GSE158723 and GSE112271) were retrieved from the Gene Expression Omnibus (GEO) database at the National Center for Biotechnology Information (NCBI) and analyzed using R software. Our analysis suggested that CAFs may promote liver cancer cell development, possibly through the interaction of pleiotrophin (PTN) and syndecan-2 (SDC2). Clinical samples from HCC patients were collected and processed into frozen sections and single-cell suspensions for Masson staining, immunofluorescence staining, and flow cytometry. Additionally, Huh7 liver cancer cells and LO2 normal liver cells were cultured and subjected to immunofluorescence assays using cell slides. Results The proportion of CAFs in cancerous tissues was higher than in adjacent non-cancerous tissues, and pleiotrophin (PTN) expression was elevated in cancer tissues compared to adjacent tissues. These findings aligned with the results of the single-cell RNA sequencing (scRNA-seq) analysis. Furthermore, SDC2 expression was significantly upregulated in Huh7 liver cancer cells compared to LO2 normal liver cells. Discussion This study suggests that CAFs may contribute to HCC progression via the PTN/SDC2 signaling pathway. Our findings provide deeper insights into the interactions between CAFs and HCC cells within the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Wenxian Lin
- Key Laboratory of Molecular Pathology for Hepatobiliary Diseases of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Department of Interventional Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Graduate School, Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Lizhu Tang
- Key Laboratory of Molecular Pathology for Hepatobiliary Diseases of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Department of Interventional Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Department of Radiation Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, The People’s Republic of China
| | - Chenyi Zhuo
- Key Laboratory of Molecular Pathology for Hepatobiliary Diseases of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
| | - Xiuli Mao
- Key Laboratory of Molecular Pathology for Hepatobiliary Diseases of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Department of Interventional Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Graduate School, Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Jiajia Shen
- Department of Laboratory Medicine, Nanning Maternity and Child Health Hospital & Nanning Women and Children’s Hospital, Nanning, 530011, The People’s Republic of China
| | - Shaoang Huang
- Graduate School, Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
| | - Shangyang Li
- Graduate School, Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Department of Laboratory Medicine, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Yujuan Qin
- Graduate School, Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Ju Liao
- Department of Interventional Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Graduate School, Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
| | - Yuhong Chen
- Department of Nephrology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, The People’s Republic of China
| | - Xiamin Zhang
- Graduate School, Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Yuting Li
- Graduate School, Guangxi University of Chinese Medicine, Nanning, 530200, The People’s Republic of China
| | - Jian Song
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Lingzhang Meng
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, Guangxi Academy of Medical Sciences & The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, The People’s Republic of China
| | - Yueyong Li
- Department of Interventional Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, The People’s Republic of China
| |
Collapse
|
11
|
Withnell E, Secrier M. SpottedPy quantifies relationships between spatial transcriptomic hotspots and uncovers environmental cues of epithelial-mesenchymal plasticity in breast cancer. Genome Biol 2024; 25:289. [PMID: 39529126 PMCID: PMC11552145 DOI: 10.1186/s13059-024-03428-y] [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: 12/15/2023] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Spatial transcriptomics is revolutionizing the exploration of intratissue heterogeneity in cancer, yet capturing cellular niches and their spatial relationships remains challenging. We introduce SpottedPy, a Python package designed to identify tumor hotspots and map spatial interactions within the cancer ecosystem. Using SpottedPy, we examine epithelial-mesenchymal plasticity in breast cancer and highlight stable niches associated with angiogenic and hypoxic regions, shielded by CAFs and macrophages. Hybrid and mesenchymal hotspot distribution follows transformation gradients reflecting progressive immunosuppression. Our method offers flexibility to explore spatial relationships at different scales, from immediate neighbors to broader tissue modules, providing new insights into tumor microenvironment dynamics.
Collapse
Affiliation(s)
- Eloise Withnell
- Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - Maria Secrier
- Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
12
|
Liu Q, Yao F, Wu L, Xu T, Na J, Shen Z, Liu X, Shi W, Zhao Y, Liao Y. Heterogeneity and interplay: the multifaceted role of cancer-associated fibroblasts in the tumor and therapeutic strategies. Clin Transl Oncol 2024; 26:2395-2417. [PMID: 38602644 DOI: 10.1007/s12094-024-03492-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/31/2024] [Indexed: 04/12/2024]
Abstract
The journey of cancer development is a multifaceted and staged process. The array of treatments available for cancer varies significantly, dictated by the disease's type and stage. Cancer-associated fibroblasts (CAFs), prevalent across various cancer types and stages, play a pivotal role in tumor genesis, progression, metastasis, and drug resistance. The strategy of concurrently targeting cancer cells and CAFs holds great promise in cancer therapy. In this review, we focus intently on CAFs, delving into their critical role in cancer's progression. We begin by exploring the origins, classification, and surface markers of CAFs. Following this, we emphasize the key cytokines and signaling pathways involved in the interplay between cancer cells and CAFs and their influence on the tumor immune microenvironment. Additionally, we examine current therapeutic approaches targeting CAFs. This article underscores the multifarious roles of CAFs within the tumor microenvironment and their potential applications in cancer treatment, highlighting their importance as key targets in overcoming drug resistance and enhancing the efficacy of tumor therapies.
Collapse
Affiliation(s)
- Qiaoqiao Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Fei Yao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Liangliang Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Tianyuan Xu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Zhen Shen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Wei Shi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
- Department of Oncology, The First Affiliated Tumor Hospital, Guangxi University of Chinese Medicine, Nanning, 530021, Guangxi, China.
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
13
|
Wei Y, Wu R, Yang S, Cao Y, Li J, Ma H, Wu J, Duan J, Yang S. MiR-137 mediated high expression of TIGD1 promotes migration, invasion, and suppresses apoptosis of lung adenocarcinoma. Lung Cancer 2024; 195:107918. [PMID: 39173230 DOI: 10.1016/j.lungcan.2024.107918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVES Tigger transposable element-derived 1 (TIGD1) expression and its underlying functions and regulatory mechanisms in lung adenocarcinoma (LUAD) remain unknown. Therefore, we intended to explore the expression, potential functions, and regulatory mechanisms of TIGD1 in LUAD. MATERIALS AND METHODS TIGD1 expression in LUAD tissues was determined by immunohistochemistry analysis of a tissue microarray. Functional experiments were conducted to determine how TIGD1 affects LUAD tumorigenesis and metastasis. The molecular mechanisms by which TIGD1 induces LUAD progression were determined. RESULTS TIGD1 was upregulated in LUAD tissues and was related to lymph node metastases. TIGD1 knockdown suppressed LUAD cell proliferation, migration, and invasion, while promoted cell apoptosis. Furthermore, decreased metastatic nodules were observed in the TIGD1 knockdown mouse metastasis model. Moreover, microarray analysis was performed to determine the potential downstream genes of TIGD1 in LUAD. Hallmark pathway analysis revealed that the downstream genes of TIGD1 were involved in epithelial-mesenchymal transition (EMT). Western blotting confirmed that vimentin and TWIST was downregulated in TIGD1 knockdown cells, while E-cadherin was upregulated. Ingenuity pathway and hallmark pathway analyses revealed that TIGD1 regulated the interleukin-6 signaling pathway and related gene members. Western blotting, quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay indicated that downregulation of TIGD1 decreased interleukin-6 and CXCL1 expression. TIGD1 expression was negatively correlated with immune infiltration in LUAD. The upstream microRNA of TIGD1 was predicted, and subsequent luciferase reporter gene experiments confirmed the interactions between miR-137 and TIGD1. The expression of miR-137 was significantly downregulated in LUAD tissues and miR-137 suppressed the proliferation, migration, and invasion of LUAD cells, partially through negatively regulating the expression of TIGD1. CONCLUSION Our findings suggest that TIGD1, which was regulated by miR-137, contributed to LUAD progression by promoting cell proliferation, migration, invasion, and EMT and suppressing cell apoptosis.
Collapse
Affiliation(s)
- Yiqun Wei
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Runmiao Wu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Yanfei Cao
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Jing Li
- Department of Traditional Chinese Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Huihui Ma
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Junfang Wu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Jinjin Duan
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Shumei Yang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| |
Collapse
|
14
|
Li SS, Liu QJ, Bao JX, Lu MT, Deng BQ, Li WW, Cao CC. Counteracting TGM2 by a Fibroin peptide ameliorated Adriamycin-induced nephropathy via regulation of lipid metabolism through PANX1-PPAR α/PANK1 pathway. Transl Res 2024; 271:26-39. [PMID: 38734063 DOI: 10.1016/j.trsl.2024.05.006] [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: 09/04/2023] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Peptide drug discovery for the treatment of chronic kidney disease (CKD) has attracted much attention in recent years due to the urge to find novel drugs and mechanisms to delay the progression of the disease. In this study, we identified a novel short peptide (named YR-7, primary sequence 'YEVEDYR') from the natural Fibroin protein, and demonstrated that it significantly alleviated pathological renal changes in ADR-induced nephropathy. PANX1 was identified as the most notably upregulated component by RNA-sequencing. Further analysis showed that YR-7 alleviated the accumulation of lipid droplets via regulation of the lipid metabolism-related proteins PPAR α and PANK1. Using chemical proteomics, fluorescence polarization, microscale thermophoresis, surface plasmon resonance, and molecular docking, YR-7 was proven to directly bind to β-barrel domains of TGM2 protein to inhibit lipid accumulation. TGM2 knockdown in vivo increased the protein levels of PPAR α and PANK1 while decreased the levels of fibrotic-related proteins to alleviate nephropathy. In vitro, overexpression TGM2 reversed the protective effects of YR-7. Co-immunoprecipitation indicated that TGM2 interacted with PANX1 to promote lipid deposition, and pharmacological inhibition or knockdown of PANX1 decreased the levels of PPAR α and PANK1 induced by ADR. Taken together, our findings revealed that TGM2-PANX1 interaction in promoting lipid deposition may be a new signaling in promoting ADR-induced nephropathy. And a novel natural peptide could ameliorate renal fibrosis through TGM2-PANX1-PPAR α/PANK1 pathway, which highlight the potential of it in the treatment of CKD.
Collapse
Affiliation(s)
- Shan-Shan Li
- Department of Nephrology, Sir Run Run hospital, Nanjing Medical University, Nanjing, China
| | - Qiao-Juan Liu
- Department of Nephrology, Sir Run Run hospital, Nanjing Medical University, Nanjing, China
| | - Jia-Xin Bao
- Department of Nephrology, Sir Run Run hospital, Nanjing Medical University, Nanjing, China
| | - Meng-Ting Lu
- Department of Nephrology, Sir Run Run hospital, Nanjing Medical University, Nanjing, China
| | - Bing-Quan Deng
- Department of Nephrology, Sir Run Run hospital, Nanjing Medical University, Nanjing, China
| | - Wen-Wen Li
- Department of Nephrology, Sir Run Run hospital, Nanjing Medical University, Nanjing, China
| | - Chang-Chun Cao
- Department of Nephrology, Sir Run Run hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Wang MJ, Zhang HL, Chen F, Guo XJ, Liu QG, Hou J. The double-edged effects of IL-6 in liver regeneration, aging, inflammation, and diseases. Exp Hematol Oncol 2024; 13:62. [PMID: 38890694 PMCID: PMC11184755 DOI: 10.1186/s40164-024-00527-1] [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: 02/24/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine and exerts its complex biological functions mainly through three different signal modes, called cis-, trans-, and cluster signaling. When IL-6 binds to its membrane or soluble receptors, the co-receptor gp130 is activated to initiate downstream signaling and induce the expression of target genes. In the liver, IL-6 can perform its anti-inflammatory activities to promote hepatocyte reprogramming and liver regeneration. On the contrary, IL-6 also exerts the pro-inflammatory functions to induce liver aging, fibrosis, steatosis, and carcinogenesis. However, understanding the roles and underlying mechanisms of IL-6 in liver physiological and pathological processes is still an ongoing process. So far, therapeutic agents against IL‑6, IL‑6 receptor (IL‑6R), IL-6-sIL-6R complex, or IL-6 downstream signal transducers have been developed, and determined to be effective in the intervention of inflammatory diseases and cancers. In this review, we summarized and highlighted the understanding of the double-edged effects of IL-6 in liver homeostasis, aging, inflammation, and chronic diseases, for better shifting the "negative" functions of IL-6 to the "beneficial" actions, and further discussed the potential therapeutic effects of targeting IL-6 signaling in the clinics.
Collapse
Affiliation(s)
- Min-Jun Wang
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Hai-Ling Zhang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Neurology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Fei Chen
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Xiao-Jing Guo
- Department of Health Statistics, Faculty of Health Service, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Qing-Gui Liu
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University (Naval Medical University), Shanghai, China.
| |
Collapse
|
16
|
Huang S, Xu Z, Zhi W, Li Y, Hu Y, Zhao F, Zhu X, Miao M, Jia Y. pH/GSH dual-responsive nanoparticle for auto-amplified tumor therapy of breast cancer. J Nanobiotechnology 2024; 22:324. [PMID: 38858692 PMCID: PMC11163783 DOI: 10.1186/s12951-024-02588-0] [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: 02/06/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
Breast cancer remains a malignancy that poses a serious threat to human health worldwide. Chemotherapy is one of the most widely effective cancer treatments in clinical practice, but it has some drawbacks such as poor targeting, high toxicity, numerous side effects, and susceptibility to drug resistance. For auto-amplified tumor therapy, a nanoparticle designated GDTF is prepared by wrapping gambogic acid (GA)-loaded dendritic porous silica nanoparticles (DPSNs) with a tannic acid (TA)-Fe(III) coating layer. GDTF possesses the properties of near-infrared (NIR)-enhanced and pH/glutathione (GSH) dual-responsive drug release, photothermal conversion, GSH depletion and hydroxyl radical (·OH) production. When GDTF is exposed to NIR laser irradiation, it can effectively inhibit cell proliferation and tumor growth both in vitro and in vivo with limited toxicity. This may be due to the synergistic effect of enhanced tumor accumulation, and elevated reactive oxygen species (ROS) production, GSH depletion, and TrxR activity reduction. This study highlights the enormous potential of auto-amplified tumor therapy.
Collapse
Affiliation(s)
- Shengnan Huang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China.
| | - Zhiling Xu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Weiwei Zhi
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Yijing Li
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China
| | - Fengqin Zhao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China
| | - Xiali Zhu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| | - Yongyan Jia
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| |
Collapse
|
17
|
Bamodu OA, Chung CC, Pisanic TR, Wu ATH. The intricate interplay between cancer stem cells and cell-of-origin of cancer: implications for therapeutic strategies. Front Oncol 2024; 14:1404628. [PMID: 38800385 PMCID: PMC11116576 DOI: 10.3389/fonc.2024.1404628] [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: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Background Cancer stem cells (CSCs) have emerged as pivotal players in tumorigenesis, disease progression, and resistance to therapies. Objective This comprehensive review delves into the intricate relationship between CSCs and the cell-of-origin in diverse cancer types. Design Comprehensive review of thematically-relevant literature. Methods We explore the underlying molecular mechanisms that drive the conversion of normal cells into CSCs and the impact of the cell-of-origin on CSC properties, tumor initiation, and therapeutic responses. Moreover, we discuss potential therapeutic interventions targeting CSCs based on their distinct cell-of-origin characteristics. Results Accruing evidence suggest that the cell-of-origin, the cell type from which the tumor originates, plays a crucial role in determining the properties of CSCs and their contribution to tumor heterogeneity. Conclusion By providing critical insights into the complex interplay between CSCs and their cellular origins, this article aims to enhance our understanding of cancer biology and pave the way for more effective and personalized cancer treatments.
Collapse
Affiliation(s)
- Oluwaseun Adebayo Bamodu
- Directorate of Postgraduate Studies, School of Clinical Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Chen-Chih Chung
- Department of Neurology, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology - Cancer Genetics and Epigenetics, Johns Hopkins University, Baltimore, MD, United States
| | - Alexander T. H. Wu
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
18
|
Røgenes H, Finne K, Winge I, Akslen LA, Östman A, Milosevic V. Development of 42 marker panel for in-depth study of cancer associated fibroblast niches in breast cancer using imaging mass cytometry. Front Immunol 2024; 15:1325191. [PMID: 38711512 PMCID: PMC11070582 DOI: 10.3389/fimmu.2024.1325191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/05/2024] [Indexed: 05/08/2024] Open
Abstract
Imaging Mass Cytometry (IMC) is a novel, and formidable high multiplexing imaging method emerging as a promising tool for in-depth studying of tissue architecture and intercellular communications. Several studies have reported various IMC antibody panels mainly focused on studying the immunological landscape of the tumor microenvironment (TME). With this paper, we wanted to address cancer associated fibroblasts (CAFs), a component of the TME very often underrepresented and not emphasized enough in present IMC studies. Therefore, we focused on the development of a comprehensive IMC panel that can be used for a thorough description of the CAF composition of breast cancer TME and for an in-depth study of different CAF niches in relation to both immune and breast cancer cell communication. We established and validated a 42 marker panel using a variety of control tissues and rigorous quantification methods. The final panel contained 6 CAF-associated markers (aSMA, FAP, PDGFRa, PDGFRb, YAP1, pSMAD2). Breast cancer tissues (4 cases of luminal, 5 cases of triple negative breast cancer) and a modified CELESTA pipeline were used to demonstrate the utility of our IMC panel for detailed profiling of different CAF, immune and cancer cell phenotypes.
Collapse
Affiliation(s)
- Hanna Røgenes
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kenneth Finne
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ingeborg Winge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Arne Östman
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Oncology and Pathology, Karolinska Institutet, Solna, Sweden
| | - Vladan Milosevic
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
19
|
Penumatsa KC, Sharma Y, Warburton RR, Singhal A, Toksoz D, Bhedi CD, Qi G, Preston IR, Anderlind C, Hill NS, Fanburg BL. Lung-specific interleukin 6 mediated transglutaminase 2 activation and cardiopulmonary fibrogenesis. Front Immunol 2024; 15:1371706. [PMID: 38650935 PMCID: PMC11033445 DOI: 10.3389/fimmu.2024.1371706] [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: 01/16/2024] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
Pulmonary hypertension (PH) pathogenesis is driven by inflammatory and metabolic derangements as well as glycolytic reprogramming. Induction of both interleukin 6 (IL6) and transglutaminase 2 (TG2) expression participates in human and experimental cardiovascular diseases. However, little is known about the role of TG2 in these pathologic processes. The current study aimed to investigate the molecular interactions between TG2 and IL6 in mediation of tissue remodeling in PH. A lung-specific IL6 over-expressing transgenic mouse strain showed elevated right ventricular (RV) systolic pressure as well as increased wet and dry tissue weights and tissue fibrosis in both lungs and RVs compared to age-matched wild-type littermates. In addition, IL6 over-expression induced the glycolytic and fibrogenic markers, hypoxia-inducible factor 1α, pyruvate kinase M2 (PKM2), and TG2. Consistent with these findings, IL6 induced the expression of both glycolytic and pro-fibrogenic markers in cultured lung fibroblasts. IL6 also induced TG2 activation and the accumulation of TG2 in the extracellular matrix. Pharmacologic inhibition of the glycolytic enzyme, PKM2 significantly attenuated IL6-induced TG2 activity and fibrogenesis. Thus, we conclude that IL6-induced TG2 activity and cardiopulmonary remodeling associated with tissue fibrosis are under regulatory control of the glycolytic enzyme, PKM2.
Collapse
Affiliation(s)
- Krishna C. Penumatsa
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, MA, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hsu CY, Mustafa MA, Kumar A, Pramanik A, Sharma R, Mohammed F, Jawad IA, Mohammed IJ, Alshahrani MY, Ali Khalil NAM, Shnishil AT, Abosaoda MK. Exploiting the immune system in hepatic tumor targeting: Unleashing the potential of drugs, natural products, and nanoparticles. Pathol Res Pract 2024; 256:155266. [PMID: 38554489 DOI: 10.1016/j.prp.2024.155266] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 04/01/2024]
Abstract
Hepatic tumors present a formidable challenge in cancer therapeutics, necessitating the exploration of novel treatment strategies. In recent years, targeting the immune system has attracted interest to augment existing therapeutic efficacy. The immune system in hepatic tumors includes numerous cells with diverse actions. CD8+ T lymphocytes, T helper 1 (Th1) CD4+ T lymphocytes, alternative M1 macrophages, and natural killer (NK) cells provide the antitumor immunity. However, Foxp3+ regulatory CD4+ T cells (Tregs), M2-like tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) are the key immune inhibitor cells. Tumor stroma can also affect these interactions. Targeting these cells and their secreted molecules is intriguing for eliminating malignant cells. The current review provides a synopsis of the immune system components involved in hepatic tumor expansion and highlights the molecular and cellular pathways that can be targeted for therapeutic intervention. It also overviews the diverse range of drugs, natural products, immunotherapy drugs, and nanoparticles that have been investigated to manipulate immune responses and bolster antitumor immunity. The review also addresses the potential advantages and challenges associated with these approaches.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 71710, Taiwan
| | | | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Atreyi Pramanik
- Institute of Pharma Sciences and Research, Chandigarh University, Mohali, India
| | - Rajiv Sharma
- Institute of Pharma Sciences and Research, Chandigarh University, Mohali, India
| | - Faraj Mohammed
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Iraq
| | | | - Imad Jasim Mohammed
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | | | | | - Munther Kadhim Abosaoda
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| |
Collapse
|
21
|
Zhang Q, Lou Y, Fang H, Sun S, Jin R, Ji Y, Chen Z. Cancer‑associated fibroblasts under therapy‑induced senescence in the tumor microenvironment (Review). Exp Ther Med 2024; 27:150. [PMID: 38476922 PMCID: PMC10928991 DOI: 10.3892/etm.2024.12438] [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: 11/14/2023] [Accepted: 01/16/2024] [Indexed: 03/14/2024] Open
Abstract
Current cancer treatments target tumor cells; however, the tumor microenvironment (TME) induces therapeutic resistance, tumor development and metastasis, thus rendering these treatments ineffective. Research on the TME has therefore concentrated on nonmalignant cells. Cancer-associated fibroblasts (CAFs) are a major TME component, which contribute to cancer progression due to their diverse origins, phenotypes and functions, including cancer cell invasion and migration, extracellular matrix remodeling, tumor metabolism modulation and therapeutic resistance. Standard cancer treatment typically exacerbates the senescence-associated secretory phenotype (SASP) of senescent cancer cells and nonmalignant cells that actively leak proinflammatory signals in the TME. Therapy-induced senescence may impair cancer cell activity and compromise treatment responsiveness. CAFs and SASP are well-studied in the formation and progression of cancer. The present review discusses the current data on CAF senescence caused by anticancer treatment and assesses how senescence-like CAFs affect tumor formation. The development of senolytic medication for aging stromal cells is also highlighted. Combining cancer therapies with senolytics may boost therapeutic effects and provide novel possibilities for research.
Collapse
Affiliation(s)
- Qiuhua Zhang
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yijie Lou
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Hao Fang
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Shaopeng Sun
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Rijuan Jin
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yunxi Ji
- Department of General Practice, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310003, P.R. China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Institute of Cancer Research, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
22
|
Zaltron E, Vianello F, Ruzza A, Palazzo A, Brillo V, Celotti I, Scavezzon M, Rossin F, Leanza L, Severin F. The Role of Transglutaminase 2 in Cancer: An Update. Int J Mol Sci 2024; 25:2797. [PMID: 38474044 DOI: 10.3390/ijms25052797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Transglutaminase type 2 (TG2) is the most ubiquitously expressed and well characterized member of the transglutaminase family. It is a ubiquitous multifunctional enzyme implicated in the regulation of several cellular pathways that support the survival, death, and general homeostasis of eukaryotic cells. Due to its multiple localizations both inside and outside the cell, TG2 participates in the regulation of many crucial intracellular signaling cascades in a tissue- and cell-specific manner, making this enzyme an important player in disease development and progression. Moreover, TG2 is capable of modulating the tumor microenvironment, a process of dynamic tissue remodeling and biomechanical events, resulting in changes which influence tumor initiation, growth, and metastasis. Even if generally related to the Ca2+-dependent post-translational modification of proteins, a number of different biological functions have been ascribed to TG2, like those of a peptide isomerase, protein kinase, guanine nucleotide binder, and cytosolic-nuclear translocator. With respect to cancer, TG2's role is controversial and highly debated; it has been described both as an anti- and pro-apoptotic factor and is linked to all the processes of tumorigenesis. However, numerous pieces of evidence support a tissue-specific role of TG2 so that it can assume both oncogenic and tumor-suppressive roles.
Collapse
Affiliation(s)
| | | | - Alessia Ruzza
- Department of Biology, University of Padua, 35131 Padua, Italy
| | - Alberta Palazzo
- Department of Biology, University of Padua, 35131 Padua, Italy
| | | | - Ilaria Celotti
- Department of Biology, University of Padua, 35131 Padua, Italy
| | | | - Federica Rossin
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Luigi Leanza
- Department of Biology, University of Padua, 35131 Padua, Italy
| | - Filippo Severin
- Department of Biology, University of Padua, 35131 Padua, Italy
| |
Collapse
|
23
|
Yao Y, Yang K, Wang Q, Zhu Z, Li S, Li B, Feng B, Tang C. Prediction of CAF-related genes in immunotherapy and drug sensitivity in hepatocellular carcinoma: a multi-database analysis. Genes Immun 2024; 25:55-65. [PMID: 38233508 PMCID: PMC10873201 DOI: 10.1038/s41435-024-00252-z] [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: 07/04/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024]
Abstract
This study aims to identify the cancer-associated fibroblasts (CAF)-related genes that can affect immunotherapy and drug sensitivity in hepatocellular carcinoma (HCC). Expression data and survival data associated with HCC were obtained in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Weighted correlation network analysis (WGCNA) analysis was performed to obtain CAF-related genes. Least Absolute Shrinkage and Selection Operator (LASSO) regression was used for regression analysis and risk models. Subsequently, Gene Set Enrichment Analysis (GSEA) analysis, Gene Set Enrichment Analysis (ssGSEA) analysis, Tumor Immune Dysfunction and Exclusion (TIDE) analysis and drug sensitivity analysis were performed on the risk models. Survival analysis of CAF scores showed that the survival rate was lower in samples with high CAF scores than those with low scores. However, this difference was not significant, suggesting CAF may not directly influence the prognosis of HCC patients. Further screening of CAF-related genes yielded 33 CAF-related genes. Seven risk models constructed based on CDR2L, SPRED1, PFKP, ENG, KLF2, FSCN1 and VCAN, showed significant differences in immunotherapy and partial drug sensitivity in HCC. Seven CAF-related genes may have important roles in immunotherapy, drug sensitivity and prognostic survival in HCC patients.
Collapse
Affiliation(s)
- Yi Yao
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - KaiQing Yang
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Qiang Wang
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Zeming Zhu
- Division 2, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Sheng Li
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Bin Li
- Division 1, Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Bin Feng
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China.
| | - Caixi Tang
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China.
| |
Collapse
|
24
|
Yao Z, Fan Y, Lin L, Kellems RE, Xia Y. Tissue transglutaminase: a multifunctional and multisite regulator in health and disease. Physiol Rev 2024; 104:281-325. [PMID: 37712623 DOI: 10.1152/physrev.00003.2023] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023] Open
Abstract
Tissue transglutaminase (TG2) is a widely distributed multifunctional protein involved in a broad range of cellular and metabolic functions carried out in a variety of cellular compartments. In addition to transamidation, TG2 also functions as a Gα signaling protein, a protein disulfide isomerase (PDI), a protein kinase, and a scaffolding protein. In the nucleus, TG2 modifies histones and transcription factors. The PDI function catalyzes the trimerization and activation of heat shock factor-1 in the nucleus and regulates the oxidation state of several mitochondrial complexes. Cytosolic TG2 modifies proteins by the addition of serotonin or other primary amines and in this way affects cell signaling. Modification of protein-bound glutamines reduces ubiquitin-dependent proteasomal degradation. At the cell membrane, TG2 is associated with G protein-coupled receptors (GPCRs), where it functions in transmembrane signaling. TG2 is also found in the extracellular space, where it functions in protein cross-linking and extracellular matrix stabilization. Of particular importance in transglutaminase research are recent findings concerning the role of TG2 in gene expression, protein homeostasis, cell signaling, autoimmunity, inflammation, and hypoxia. Thus, TG2 performs a multitude of functions in multiple cellular compartments, making it one of the most versatile cellular proteins. Additional evidence links TG2 with multiple human diseases including preeclampsia, hypertension, cardiovascular disease, organ fibrosis, cancer, neurodegenerative diseases, and celiac disease. In conclusion, TG2 provides a multifunctional and multisite response to physiological stress.
Collapse
Affiliation(s)
- Zhouzhou Yao
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yuhua Fan
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Lizhen Lin
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School at Houston, Houston, Texas, United States
| | - Yang Xia
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
25
|
Liu S, Xu W, Shu H, Dai Y, Du Y, Liu Y, Huang L, Sun G. Associations of circulating immunomarkers with the efficacy of immunotherapy for primary hepatic carcinoma. Cancer Med 2023; 12:21830-21848. [PMID: 38054365 PMCID: PMC10757102 DOI: 10.1002/cam4.6754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/29/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Peripheral blood immunomarkers are associated with prognosis in patients with solid tumors receiving chemotherapy or immunotherapy. In this study, the associations of circulating neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), and platelet-to-lymphocyte ratio (PLR), as well as their dynamic changes were investigated in relation to the efficacy of immunotherapy in patients with primary liver cancer. METHODS Comparisons were made between NLR, MLR, and PLR among individuals exhibiting disease control (defined as the best response of partial response [PR] or stable disease [SD]) and those with progressive disease (PD). Additionally, disease control rate (DCR), overall survival (OS), and progression-free survival (PFS) were compared between individuals with different NLR, MLR, and PLR levels before initiating palliative immunotherapy. Furthermore, comparisons were made between patients with different alterations in the ratios at the second cycle of immunotherapy compared to baseline. These analyses were performed using univariate and multivariate approaches. A total of 119 Chinese patients with liver cancer who underwent immunotherapy were included in this study, which focused on hepatocellular carcinoma (HCC). RESULTS In cases with HCC (n = 104), the cutoffs of NLR, MLR, and PLR to differentiate treatment responders from nonresponders were 3.38, 0.28, and 227.18, respectively. Patients with the best response of PR or SD had significantly lower NLR and MLR. Patients with NLR <3.38 and those with MLR <0.28 significantly had longer OS and PFS than their counterparts, and those with PLR <227.18 had significantly longer PFS, both in overall patients and in various patient subgroups. Lower NLR, MLR, or PLR was associated with earlier BCLC stage, fewer metastatic sites, less frequent extrahepatic metastasis, or better performance status. For individuals who had an unfavorable baseline NLR ≥3.38, MLR ≥0.28, or a favorable baseline PLR <227.18 prior to first immunotherapy, a decrease in NLR, MLR, or PLR at Cycle 2 of immunotherapy was significantly associated with a higher DCR. CONCLUSIONS Among patients with HCC who received immunotherapy, lower NLR, and MLR at baseline in overall patients were significantly associated with better disease control and more favorable survival outcomes (both OS and PFS), and lower PLR was significantly associated with longer PFS. The findings of this research may offer useful hints foranoptimized selection of patients with liver cancer who may benefit more from immunotherapy.
Collapse
Affiliation(s)
- Sha Liu
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Wentao Xu
- School of Clinical MedicineAnhui Medical UniversityHefeiAnhuiChina
| | - Hang Shu
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Ying Dai
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Yingying Du
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Yunmei Liu
- School of Cultural Heritage and Information ManagementShanghai UniversityShanghaiChina
| | - Lei Huang
- Department of Oncology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Medical Center on Aging of Ruijin Hospital, MCARJHShanghai Jiaotong University School of MedicineShanghaiChina
| | - Guoping Sun
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
26
|
Arvanitakis K, Papadakos SP, Lekakis V, Koufakis T, Lempesis IG, Papantoniou E, Kalopitas G, Georgakopoulou VE, Stergiou IE, Theocharis S, Germanidis G. Meeting at the Crossroad between Obesity and Hepatic Carcinogenesis: Unique Pathophysiological Pathways Raise Expectations for Innovative Therapeutic Approaches. Int J Mol Sci 2023; 24:14704. [PMID: 37834153 PMCID: PMC10572430 DOI: 10.3390/ijms241914704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
The escalating global prevalence of obesity and its intricate association with the development of hepatocellular carcinoma (HCC) pose a substantial challenge to public health. Obesity, acknowledged as a pervasive epidemic, is linked to an array of chronic diseases, including HCC, catalyzing the need for a comprehensive understanding of its molecular underpinnings. Notably, HCC has emerged as a leading malignancy with rising incidence and mortality. The transition from viral etiologies to the prominence of metabolic dysfunction-associated fatty liver disease (MAFLD)-related HCC underscores the urgent need to explore the intricate molecular pathways linking obesity and hepatic carcinogenesis. This review delves into the interwoven landscape of molecular carcinogenesis in the context of obesity-driven HCC while also navigating using the current therapeutic strategies and future prospects for combating obesity-related HCC. We underscore the pivotal role of obesity as a risk factor and propose an integrated approach encompassing lifestyle interventions, pharmacotherapy, and the exploration of emerging targeted therapies. As the obesity-HCC nexus continues to challenge healthcare systems globally, a comprehensive understanding of the intricate molecular mechanisms and innovative therapeutic strategies is imperative to alleviate the rising burden of this dual menace.
Collapse
Affiliation(s)
- Konstantinos Arvanitakis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.A.); (E.P.); (G.K.)
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stavros P. Papadakos
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (S.T.)
| | - Vasileios Lekakis
- Department of Gastroenterology, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece;
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Centre, First Department of Internal Medicine, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Ioannis G. Lempesis
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Eleni Papantoniou
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.A.); (E.P.); (G.K.)
| | - Georgios Kalopitas
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.A.); (E.P.); (G.K.)
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | | | - Ioanna E. Stergiou
- Pathophysiology Department, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Stamatios Theocharis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (S.T.)
| | - Georgios Germanidis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.A.); (E.P.); (G.K.)
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| |
Collapse
|
27
|
Ma Z, Sun Q, Zhang C, Zheng Q, Liu Y, Xu H, He Y, Yao C, Chen J, Xia H. RHOJ Induces Epithelial-to-Mesenchymal Transition by IL-6/STAT3 to Promote Invasion and Metastasis in Gastric Cancer. Int J Biol Sci 2023; 19:4411-4426. [PMID: 37781036 PMCID: PMC10535698 DOI: 10.7150/ijbs.81972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 07/20/2023] [Indexed: 10/03/2023] Open
Abstract
Background: Recently, the molecular classification of gastric cancer (GC) promotes the advances of GC patients' precision therapy and prognosis prediction. According to the Asian Cancer Research Group (ACRG), GC is classified as microsatellite instable (MSI) subtype GC, microsatellite stable/epithelial-to-mesenchymal transition (MSS/EMT) subtype GC, MSS/TP53- subtype GC, and MSS/TP53+ subtype GC. Due to the easy metastasis of EMT-subtype GC, it has the worst prognosis, the highest recurrence rate, and the tendency to occur at a younger age. Therefore, it is curious and crucial for us to understand the molecular basis of EMT-subtype GC. Methods: The expression of RHOJ was detected by quantitative real-time PCR (qPCR) and immunohistochemistry (IHC) in GC cells and tissues. Western blotting and immunofluorescence (IF) were conducted to examine the effects of RHOJ on the EMT markers' expression of GC cells. The GC cells' migration and invasion were investigated by transwell assay. The tumor growth and metastasis were demonstrated correspondingly in different xenograft models. Results: Firstly, it was noticed that RHOJ was significantly upregulated in EMT-subtype GC and RHOJ has close relationships with the EMT process of GC, based on the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) databases. Next, transwell assay and tail vein metastasis models were conducted to verify that RHOJ mediates the EMT to regulate the invasion and metastasis of GC in vitro and in vivo. In addition, weakened tumor angiogenesis was observed after RHOJ knockdown by the angiogenesis assay of HUVEC. RNA-seq and further study unveiled that RHOJ aggravates the malignant progression of GC by inducing EMT through IL-6/STAT3 to promote invasion and metastasis. Finally, blocking the IL-6/STAT3 signaling overcame RHOJ-mediated GC cells' growth and migration. Conclusions: These results indicate that the upregulation of RHOJ contributes to EMT-subtype GC invasion and metastasis via IL-6/STAT3 signaling, and RHOJ is expected to become a promising biomarker and therapeutic target for EMT-subtype GC patients.
Collapse
Affiliation(s)
- Zhijie Ma
- Zhongda Hospital, School of Medicine & Advanced Institute for Life and Health, Southeast University, Nanjing 210009, China
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, China
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Qi Sun
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Chengfei Zhang
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Qian Zheng
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Yixuan Liu
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Haojun Xu
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Yiting He
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Chengyun Yao
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Jinfei Chen
- Department of Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang, China
| | - Hongping Xia
- Zhongda Hospital, School of Medicine & Advanced Institute for Life and Health, Southeast University, Nanjing 210009, China
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, China
- School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
28
|
Mottais A, Riberi L, Falco A, Soccal S, Gohy S, De Rose V. Epithelial-Mesenchymal Transition Mechanisms in Chronic Airway Diseases: A Common Process to Target? Int J Mol Sci 2023; 24:12412. [PMID: 37569787 PMCID: PMC10418908 DOI: 10.3390/ijms241512412] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a reversible process, in which epithelial cells lose their epithelial traits and acquire a mesenchymal phenotype. This transformation has been described in different lung diseases, such as lung cancer, interstitial lung diseases, asthma, chronic obstructive pulmonary disease and other muco-obstructive lung diseases, such as cystic fibrosis and non-cystic fibrosis bronchiectasis. The exaggerated chronic inflammation typical of these pulmonary diseases can induce molecular reprogramming with subsequent self-sustaining aberrant and excessive profibrotic tissue repair. Over time this process leads to structural changes with progressive organ dysfunction and lung function impairment. Although having common signalling pathways, specific triggers and regulation mechanisms might be present in each disease. This review aims to describe the various mechanisms associated with fibrotic changes and airway remodelling involved in chronic airway diseases. Having better knowledge of the mechanisms underlying the EMT process may help us to identify specific targets and thus lead to the development of novel therapeutic strategies to prevent or limit the onset of irreversible structural changes.
Collapse
Affiliation(s)
- Angélique Mottais
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
| | - Luca Riberi
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Andrea Falco
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Simone Soccal
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Cystic Fibrosis Reference Centre, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Virginia De Rose
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
29
|
Liu Y, Chen X, Luo W, Zhao Y, Nashan B, Huang L, Yuan X. Identification and validation of Birc5 as a novel activated cell cycle program biomarker associated with infiltration of immunosuppressive myeloid-derived suppressor cells in hepatocellular carcinoma. Cancer Med 2023; 12:16370-16385. [PMID: 37326143 PMCID: PMC10469657 DOI: 10.1002/cam4.6271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Preclinical studies and clinical trials have demonstrated that tumor-intrinsic activation of the cell cycle program impedes anticancer immunotherapy. Identification of cell cycle-related biomarkers may provide novel therapeutic targets to augment the efficacy of immunotherapy in hepatocellular carcinoma (HCC). METHOD AND RESULTS Based on the genes related to cell cycle program, two clusters (Cluster 1 and Cluster 2) were detected in HCC patients via non-negative matrix factorization algorithm. Multivariable-adjusted Cox regression analysis indicated that the cell cycle gene-based classification was a significant prognostic factor for predicting the clinical outcome of HCC patients. Cluster 1 showed shorter overall survival time and progression-free interval time was associated with activated cell cycle program, higher infiltration of myeloid-derived suppressor cells (MDSCs) and less sensitivity to immunotherapy. A three-gene prognostic model, including BIRC5, C8G, and SPP1, was constructed to characterize the cell cycle-based classification of HCC, which had strong robustness and a stable predictive performance. Notably, Birc5 was positively correlated with CD11b expression (a MDSC marker) in HCC tissue. Concordant high expression of Birc5 and intratumor infiltration level of MDSCs were correlated with worse prognosis of HCC patients. In vitro, hepatocellular Birc5 overexpression promoted immunosuppressive CD11b+ CD33+ HLA-DR- MDSC expansion from human peripheral blood mononuclear cells. Genetically modified animal model of liver cancer revealed that Birc5 depletion upregulated the genes related to lymphocyte-mediated immunity, natural killer cell-mediated immunity, interferon-gamma production, T-cell activation, and T-cell-mediated cytotoxicity. These results suggest an immunosuppressive function of Birc5 in HCC. CONCLUSION Birc5 was a potential biomarker and inducer of intratumor infiltration of MDSCs, which led to T cell exclusion or dysfunction in tumor immune microenvironment, consequently resulting in reduced response to ICIs in HCC.
Collapse
Affiliation(s)
- Yun Liu
- Department of Radiation Oncology, Anhui Provincial Cancer HospitalThe First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Xi Chen
- Department of Gastrointestinal Oncology Surgery, Anhui Provincial Cancer HospitalThe First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Wenwu Luo
- Department of PathologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Yufei Zhao
- Department of Radiation Oncology, Anhui Provincial Cancer HospitalThe First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Björn Nashan
- Organ Transplant Center, Department of Hepatobiliary and Transplantation Surgery, The First Affiliated Hospital of USTCDivision of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Lei Huang
- Department of OncologyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Medical Center on Aging of Ruijin Hospital, MCARJH, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Xiaodong Yuan
- Organ Transplant Center, Department of Hepatobiliary and Transplantation Surgery, The First Affiliated Hospital of USTCDivision of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| |
Collapse
|
30
|
Bates ME, Libring S, Reinhart-King CA. Forces exerted and transduced by cancer-associated fibroblasts during cancer progression. Biol Cell 2023; 115:e2200104. [PMID: 37224184 PMCID: PMC10757454 DOI: 10.1111/boc.202200104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/13/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
Although it is well-known that cancer-associated fibroblasts (CAFs) play a key role in regulating tumor progression, the effects of mechanical tissue changes on CAFs are understudied. Myofibroblastic CAFs (myCAFs), in particular, are known to alter tumor matrix architecture and composition, heavily influencing the mechanical forces in the tumor microenvironment (TME), but much less is known about how these mechanical changes initiate and maintain the myCAF phenotype. Additionally, recent studies have pointed to the existence of CAFs in circulating tumor cell clusters, indicating that CAFs may be subject to mechanical forces beyond the primary TME. Due to their pivotal role in cancer progression, targeting CAF mechanical regulation may provide therapeutic benefit. Here, we will discuss current knowledge and summarize existing gaps in how CAFs regulate and are regulated by matrix mechanics, including through stiffness, solid and fluid stresses, and fluid shear stress.
Collapse
Affiliation(s)
- Madison E Bates
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Sarah Libring
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | | |
Collapse
|
31
|
Sun B, Lei X, Cao M, Li Y, Yang LY. Hepatocellular carcinoma cells remodel the pro-metastatic tumour microenvironment through recruitment and activation of fibroblasts via paracrine Egfl7 signaling. Cell Commun Signal 2023; 21:180. [PMID: 37480091 PMCID: PMC10362567 DOI: 10.1186/s12964-023-01200-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/16/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND The tumour microenvironment consists of a complex and dynamic milieu of cancer cells, including tumour-associated stromal cells (leukocytes, fibroblasts, vascular cells, etc.) and their extracellular products. During invasion and metastasis, cancer cells actively remodel the tumour microenvironment and alterations of microenvironment, particularly cancer-associated fibroblasts (CAFs), can promote tumour progression. However, the underlying mechanisms of the CAF formation and their metastasis-promoting functions remain unclear. METHODS Primary liver fibroblasts and CAFs were isolated and characterized. CAFs in clinical samples were identified by immunohistochemical staining and the clinical significance of CAFs was also analysed in two independent cohorts. A transwell coculture system was used to confirm the role of HCC cells in CAF recruitment and activation. qRT-PCR, western blotting and ELISA were used to screen paracrine cytokines. The role and mechanism of Egfl7 in CAFs were explored via an in vitro coculture system and an in vivo mouse orthotopic transplantation model. RESULTS We showed that CAFs in hepatocellular carcinoma (HCC) are characterized by the expression of α-SMA and that HCC cells can recruit liver fibroblasts (LFs) and activate them to promote their transformation into CAFs. High α-SMA expression, indicating high CAF infiltration, was correlated with malignant characteristics. It was also an independent risk factor for HCC survival and could predict a poor prognosis in HCC patients. Then, we demonstrated that EGF-like domain multiple 7 (Egfl7) was preferentially secreted by HCC cells, and exhibited high potential to recruit and activate LFs into the CAF phenotype. The ability of Egfl7 to modulate LFs relies upon increased phosphorylation of FAK and AKT via the receptor ανβ3 integrin. Strikingly, CAFs activated by paracrine Egfl7 could further remodel the tumour microenvironment by depositing fibrils and collagen and in turn facilitate HCC cell proliferation, invasion and metastasis. CONCLUSION Our data highlighted a novel role of Egfl7 in remodelling the tumour microenvironment: it recruits LFs and activates them to promote their transformation into CAFs via the ανβ3 integrin signaling pathway, which further promotes HCC progression and contributes to poor clinical outcomes in HCC patients. Video Abstract.
Collapse
Affiliation(s)
- Bo Sun
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Xiong Lei
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, China
| | - Momo Cao
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, China
| | - Yiming Li
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, China
| | - Lian-Yue Yang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, China.
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
32
|
Monleón-Guinot I, Milian L, Martínez-Vallejo P, Sancho-Tello M, Llop-Miguel M, Galbis JM, Cremades A, Carda C, Mata M. Morphological Characterization of Human Lung Cancer Organoids Cultured in Type I Collagen Hydrogels: A Histological Approach. Int J Mol Sci 2023; 24:10131. [PMID: 37373279 DOI: 10.3390/ijms241210131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/03/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The malignity of lung cancer is conditioned by the tumor microenvironment (TME), in which cancer-associated fibroblasts (CAFs) are relevant. In this work, we generated organoids by combining A549 cells with CAFs and normal fibroblasts (NF) isolated from adenocarcinoma tumors. We optimized the conditions for their manufacture in a short time. We evaluated the morphology of organoids using confocal microscopy analysis of F-actin, vimentin and pankeratin. We determined the ultrastructure of the cells in the organoids via transmission electron microscopy and the expression of CDH1, CDH2 and VIM via RT-PCR. The addition of stromal cells induces the self-organization of the organoids, which acquired a bowl morphology, as well as their growth and the generation of cell processes. They also influenced the expression of genes related to epithelial mesenchymal transition (EMT). CAFs potentiated these changes. All cells acquired a characteristic secretory phenotype, with cohesive cells appearing inside the organoids. In the periphery, many cells acquired a migratory phenotype, especially in organoids that incorporated CAFs. The deposit of abundant extracellular matrix could also be observed. The results presented here reinforce the role of CAFs in the progression of lung tumors and could lay the foundation for a useful in vitro pharmacological model.
Collapse
Affiliation(s)
- Irene Monleón-Guinot
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Lara Milian
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Patricia Martínez-Vallejo
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
| | - María Sancho-Tello
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Mauro Llop-Miguel
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
| | | | - Antonio Cremades
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
- Hospital de la Ribera, 46600 Alzira, Spain
| | - Carmen Carda
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Manuel Mata
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- Biomedical Research Networking Center of Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
33
|
Nigam M, Mishra AP, Deb VK, Dimri DB, Tiwari V, Bungau SG, Bungau AF, Radu AF. Evaluation of the association of chronic inflammation and cancer: Insights and implications. Biomed Pharmacother 2023; 164:115015. [PMID: 37321055 DOI: 10.1016/j.biopha.2023.115015] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/02/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023] Open
Abstract
Among the most extensively researched processes in the development and treatment of cancer is inflammatory condition. Although acute inflammation is essential for the wound healing and reconstruction of tissues that have been damaged, chronic inflammation may contribute to the onset and growth of a number of diseases, including cancer. By disrupting the signaling processes of cells, which result in cancer induction, invasion, and development, a variety of inflammatory molecules are linked to the development of cancer. The microenvironment surrounding the tumor is greatly influenced by inflammatory cells and their subsequent secretions, which also contribute significantly to the tumor's growth, survivability, and potential migration. These inflammatory variables have been mentioned in several publications as prospective diagnostic tools for anticipating the onset of cancer. Targeting inflammation with various therapies can reduce the inflammatory response and potentially limit or block the proliferation of cancer cells. The scientific medical literature from the past three decades has been studied to determine how inflammatory chemicals and cell signaling pathways related to cancer invasion and metastasis are related. The current narrative review updates the relevant literature while highlighting the specifics of inflammatory signaling pathways in cancer and their possible therapeutic possibilities.
Collapse
Affiliation(s)
- Manisha Nigam
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, 246174 Srinagar Garhwal, Uttarakhand, India
| | - Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, 9300 Bloemfontein, South Africa.
| | - Vishal Kumar Deb
- Dietetics and Nutrition Technology Division, CSIR Institute of Himalayan Bioresource Technology, 176061 Palampur, Himanchal Pradesh, India
| | - Deen Bandhu Dimri
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, 246174 Srinagar Garhwal, Uttarakhand, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi 221005, Uttar Pradesh, India
| | - Simona Gabriela Bungau
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Alexa Florina Bungau
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Andrei-Flavius Radu
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
34
|
Del Nero M, Colombo A, Garbujo S, Baioni C, Barbieri L, Innocenti M, Prosperi D, Colombo M, Fiandra L. Advanced Cell Culture Models Illuminate the Interplay between Mammary Tumor Cells and Activated Fibroblasts. Cancers (Basel) 2023; 15:cancers15092498. [PMID: 37173963 PMCID: PMC10177476 DOI: 10.3390/cancers15092498] [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: 02/28/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The interaction between tumor cells and activated fibroblasts determines malignant features of desmoplastic carcinomas such as rapid growth, progression towards a metastatic phenotype, and resistance to chemotherapy. On one hand, tumor cells can activate normal fibroblasts and even reprogram them into CAFs through complex mechanisms that also involve soluble factors. Among them, transforming growth factor beta (TGF-β) and Platelet-Derived Growth Factor (PDGF) have an established role in the acquisition of pro-tumorigenic phenotypes by fibroblasts. On the other hand, activated fibroblasts release Interleukin-6 (IL-6), which increases tumor-cell invasiveness and chemoresistance. However, the interplay between breast cancer cells and fibroblasts, as well as the modes of action of TGF-β, PDGF, and IL-6, are difficult to investigate in vivo. Here, we validated the usage of advanced cell culture models as tools to study the interplay between mammary tumor cells and fibroblasts, taking mouse and human triple-negative tumor cells and fibroblasts as a case study. We employed two different settings, one permitting only paracrine signaling, the other both paracrine and cell-contact-based signaling. These co-culture systems allowed us to unmask how TGF-β, PDGF and IL-6 mediate the interplay between mammary tumor cells and fibroblasts. We found that the fibroblasts underwent activation induced by the TGF-β and the PDGF produced by the tumor cells, which increased their proliferation and IL-6 secretion. The IL-6 secreted by activated fibroblasts enhanced tumor-cell proliferation and chemoresistance. These results show that these breast cancer avatars possess an unexpected high level of complexity, which resembles that observed in vivo. As such, advanced co-cultures provide a pathologically relevant tractable system to study the role of the TME in breast cancer progression with a reductionist approach.
Collapse
Affiliation(s)
- Martina Del Nero
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Alessandro Colombo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Stefania Garbujo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Chiara Baioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Linda Barbieri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Metello Innocenti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Davide Prosperi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Miriam Colombo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Luisa Fiandra
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
35
|
Ma XL, Nie YY, Xie SH, Zheng H, Tong Y, Wang YC, Yan TQ, Meng X, Cao JZ, Tang WG, Guo L, Lu RQ. ASAP2 interrupts c-MET-CIN85 interaction to sustain HGF/c-MET-induced malignant potentials in hepatocellular carcinoma. Exp Hematol Oncol 2023; 12:38. [PMID: 37061723 PMCID: PMC10105420 DOI: 10.1186/s40164-023-00393-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/27/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Sustained activation of hepatocyte growth factor (HGF)/c-MET signaling is a major driver of hepatocellular carcinoma (HCC) progression, but underlying mechanism is unclear. ArfGAP With SH3 Domain, Ankyrin Repeat And PH Domain 2 (ASAP2) can reportedly activate GTPases and promote receptor tyrosine kinase signaling. However, the exact role of ASAP2 in HCC, especially for c-MET activation, also remains elusive. METHODS ASAP2 expression levels in HCC tissues and cells were quantified using qRT-PCR, western blot (WB) analysis, and immunohistochemistry staining. Cell counting kit-8 (CCK-8) and colony formation assays were performed to evaluate cell proliferation rates. Flow cytometry assays were conducted to assess apoptosis rates. Wound healing and Transwell assays were performed to determine cell migration and invasion capacities. Epithelial-mesenchymal transition (EMT)-related marker expression levels were also examined. Subcutaneous implantation and tail vein injection models were applied for in vivo growth and metastasis evaluations, respectively. Bioinformatics analyses of The Cancer Genome Atlas and STRING datasets were performed to explore ASAP2 downstream signaling. Co-immunoprecipitation and Cycloheximide chasing experiments were performed to assess protein-protein interactions and protein half-life, respectively. RESULTS ASAP2 had higher expression levels in HCC tissues than in normal liver, and also predicted poor prognosis. Knocking down ASAP2 significantly impaired cell proliferation, migration, and invasion capacities, but promoted apoptosis in HCC cells in vitro. However, overexpression of ASAP2 achieved the opposite effects. In vivo experiments confirmed that ASAP2 could promote HCC cell growth and facilitate lung metastasis. Interestingly, ASAP2 was essential for triggering EMT. Gene Set Enrichment Analysis demonstrated that c-MET signaling was greatly enriched in ASAP2-high HCC cases. Additionally, c-MET signaling activity was significantly decreased following ASAP knockdown, evidenced by reduced c-MET, p-AKT, and p-ERK1/2 protein levels. Importantly, ASAP2 knockdown effectively attenuated HGF/c-MET signaling-induced malignant phenotypes. c-MET and ASAP2 expression levels were positively correlated in our cohort. Mechanistically, ASAP2 can directly bind to CIN85, thereby disrupting its interaction with c-MET, and can thus antagonize CIN85-induced c-MET internalization and lysosome-mediated degradation. Notably, knocking down CIN85 can rescue the observed inhibitory effects caused by ASAP2 knockdown. CONCLUSIONS This study highlights the importance of ASAP2 in sustaining c-MET signaling, which can facilitate HCC progression.
Collapse
Affiliation(s)
- Xiao-Lu Ma
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yan-Yan Nie
- Shanghai Lab. Animal Research Center, Shanghai, 201203, China
| | - Su-Hong Xie
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Hui Zheng
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Ying Tong
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Yan-Chun Wang
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Tian-Qing Yan
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Xin Meng
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Jia-Zhen Cao
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China
| | - Wei-Guo Tang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Lin Guo
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| | - Ren-Quan Lu
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
36
|
Starska-Kowarska K. The Role of Different Immunocompetent Cell Populations in the Pathogenesis of Head and Neck Cancer-Regulatory Mechanisms of Pro- and Anti-Cancer Activity and Their Impact on Immunotherapy. Cancers (Basel) 2023; 15:1642. [PMID: 36980527 PMCID: PMC10046400 DOI: 10.3390/cancers15061642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/10/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most aggressive and heterogeneous groups of human neoplasms. HNSCC is characterized by high morbidity, accounting for 3% of all cancers, and high mortality with ~1.5% of all cancer deaths. It was the most common cancer worldwide in 2020, according to the latest GLOBOCAN data, representing the seventh most prevalent human malignancy. Despite great advances in surgical techniques and the application of modern combinations and cytotoxic therapies, HNSCC remains a leading cause of death worldwide with a low overall survival rate not exceeding 40-60% of the patient population. The most common causes of death in patients are its frequent nodal metastases and local neoplastic recurrences, as well as the relatively low response to treatment and severe drug resistance. Much evidence suggests that the tumour microenvironment (TME), tumour infiltrating lymphocytes (TILs) and circulating various subpopulations of immunocompetent cells, such regulatory T cells (CD4+CD25+Foxp3+Tregs), cytotoxic CD3+CD8+ T cells (CTLs) and CD3+CD4+ T helper type 1/2/9/17 (Th1/Th2/Th9/Th17) lymphocytes, T follicular helper cells (Tfh) and CD56dim/CD16bright activated natural killer cells (NK), carcinoma-associated fibroblasts (CAFs), myeloid-derived suppressor cells (MDSCs), tumour-associated neutrophils (N1/N2 TANs), as well as tumour-associated macrophages (M1/M2 phenotype TAMs) can affect initiation, progression and spread of HNSCC and determine the response to immunotherapy. Rapid advances in the field of immuno-oncology and the constantly growing knowledge of the immunosuppressive mechanisms and effects of tumour cancer have allowed for the use of effective and personalized immunotherapy as a first-line therapeutic procedure or an essential component of a combination therapy for primary, relapsed and metastatic HNSCC. This review presents the latest reports and molecular studies regarding the anti-tumour role of selected subpopulations of immunocompetent cells in the pathogenesis of HNSCC, including HPV+ve (HPV+) and HPV-ve (HPV-) tumours. The article focuses on the crucial regulatory mechanisms of pro- and anti-tumour activity, key genetic or epigenetic changes that favour tumour immune escape, and the strategies that the tumour employs to avoid recognition by immunocompetent cells, as well as resistance mechanisms to T and NK cell-based immunotherapy in HNSCC. The present review also provides an overview of the pre- and clinical early trials (I/II phase) and phase-III clinical trials published in this arena, which highlight the unprecedented effectiveness and limitations of immunotherapy in HNSCC, and the emerging issues facing the field of HNSCC immuno-oncology.
Collapse
Affiliation(s)
- Katarzyna Starska-Kowarska
- Department of Physiology, Pathophysiology and Clinical Immunology, Department of Clinical Physiology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland; ; Tel.: +48-604-541-412
- Department of Otorhinolaryngology, EnelMed Center Expert, Drewnowska 58, 91-001 Lodz, Poland
| |
Collapse
|
37
|
Wu Y, Clark KC, Niranjan B, Chüeh AC, Horvath LG, Taylor RA, Daly RJ. Integrative characterisation of secreted factors involved in intercellular communication between prostate epithelial or cancer cells and fibroblasts. Mol Oncol 2023; 17:469-486. [PMID: 36608258 PMCID: PMC9980303 DOI: 10.1002/1878-0261.13376] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/05/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023] Open
Abstract
Reciprocal interactions between prostate cancer cells and carcinoma-associated fibroblasts (CAFs) mediate cancer development and progression; however, our understanding of the signalling pathways mediating these cellular interactions remains incomplete. To address this, we defined secretome changes upon co-culture of prostate epithelial or cancer cells with fibroblasts that mimic bi-directional communication in tumours. Using antibody arrays, we profiled conditioned media from mono- and co-cultures of prostate fibroblasts, epithelial and cancer cells, identifying secreted proteins that are upregulated in co-culture compared to mono-culture. Six of these (CXCL10, CXCL16, CXCL6, FST, PDGFAA, IL-17B) were functionally screened by siRNA knockdown in prostate cancer cell/fibroblast co-cultures, revealing a key role for follistatin (FST), a secreted glycoprotein that binds and bioneutralises specific members of the TGF-β superfamily, including activin A. Expression of FST by both cell types was required for the fibroblasts to enhance prostate cancer cell proliferation and migration, whereas FST knockdown in co-culture grafts decreased tumour growth in mouse xenografts. This study highlights the complexity of prostate cancer cell-fibroblast communication, demonstrates that co-culture secretomes cannot be predicted from individual cultures, and identifies FST as a tumour-microenvironment-derived secreted factor that represents a candidate therapeutic target.
Collapse
Affiliation(s)
- Yunjian Wu
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVictoriaAustralia
| | - Kimberley C. Clark
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVictoriaAustralia
| | - Birunthi Niranjan
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Department of Anatomy and Developmental BiologyMonash UniversityClaytonVictoriaAustralia
| | - Anderly C. Chüeh
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVictoriaAustralia
| | - Lisa G. Horvath
- Garvan Institute of Medical ResearchDarlinghurstNew South WalesAustralia
- University of SydneyNew South WalesAustralia
- Chris O'Brien LifehouseSydneyNew South WalesAustralia
| | - Renea A. Taylor
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Department of PhysiologyMonash UniversityClaytonVictoriaAustralia
- Cancer Research Division, Peter MacCallum Cancer CentreThe University of MelbourneVictoriaAustralia
| | - Roger J. Daly
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
38
|
Li X, Ma Y, Wu J, Ni M, Chen A, Zhou Y, Dai W, Chen Z, Jiang R, Ling Y, Yao Q, Chen W. Thiol oxidative stress-dependent degradation of transglutaminase2 via protein S-glutathionylation sensitizes 5-fluorouracil therapy in 5-fluorouracil-resistant colorectal cancer cells. Drug Resist Updat 2023; 67:100930. [PMID: 36736043 DOI: 10.1016/j.drup.2023.100930] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
5-Fluorouracil (5-Fu) is a first-line drug for colorectal cancer (CRC) therapy. However, the development of 5-Fu resistance limits its chemotherapeutic effectiveness and often leads to poor prognoses of CRC. Transglutaminase 2 (TGM2), a member of the transglutaminase family, is considered to be associated with chemoresistance through apoptotic prevention in various cancers including CRC. TGM2 was found to be overexpressed in two 5-Fu-resistant CRC cell lines and down-regulated by increased thiol oxidative stress induced by inhibition of glutathione reductase (GR). The present study aimed to explore the role of TGM2 in 5-Fu-resistant CRC and the mechanism of action by which the elevated thiol oxidative stress down-regulates TGM2 protein level. The results revealed that 5-Fu-resistance induced by overexpression of TGM2 in CRC cells was reversed through up-regulation of thiol oxidative stress. Knockdown of TGM2 increased the chemosensitivity of CRC cells to 5-Fu. Thiol oxidative stress potentially enhanced the therapeutic effect of 5-Fu in the resistant CRC cells by promotion of 5-Fu-induced apoptosis through down-regulation of TGM2. The elevated thiol oxidative stress increased the S-glutathionylation of TGM2 and led to proteasomal degradation of TGM2. Furthermore, Cys193 was identified as the S-glutathionylation site in TGM2, and its mutation resulted in thiol oxidative stress-mediated CRC cell apoptotic resistance. TGM2-induced EMT was also suppressed by the elevated thiol oxidative stress. A xenograft tumor model confirmed the effect of thiol oxidative stress in the reversal of 5-Fu resistance in CRC cells in vivo. TGM2 protein expression level was found to be significantly higher in human CRC specimens than in non-cancerous colorectal tissues. Taken together, the present data suggest an important role of TGM2 in 5-Fu resistance in CRC cells. Up-regulation of thiol oxidative stress could be a potential therapeutic approach for treating 5-Fu-resistant CRC and TGM2 may serve as a potential therapeutic target of thiol oxidative stress.
Collapse
Affiliation(s)
- Xia Li
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine on Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yan Ma
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Junzhou Wu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Maowei Ni
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Aiping Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yun Zhou
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Wumin Dai
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Zhongjian Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ruibin Jiang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
| | - Yutian Ling
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Qinghua Yao
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Department of Integrated Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine on Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Wei Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine on Cancer, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
39
|
Zhang Q, Wang Y, Liu F. Cancer-associated fibroblasts: Versatile mediators in remodeling the tumor microenvironment. Cell Signal 2023; 103:110567. [PMID: 36538999 DOI: 10.1016/j.cellsig.2022.110567] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Current cancer therapeutic strategies are generally not sufficient to eradicate malignancy, as cancer stroma cells contribute to tumor evasion and therapeutic resistance. Cancer-associated fibroblasts (CAFs) constitute a largely heterogeneous type of stromal cell population and are important components of the tumor microenvironment (TME). CAFs are the most abundant stromal cell type and are actively involved in tumor progression through complex mechanisms involving effects on other cell types. Research conducted in recent years has emphasized an emerging function of CAFs in the remodeling of the TME that promotes tumor progression with effects on response to treatment by various molecular mechanisms. A comprehensive mechanism of tumor-promoting activities of CAFs could facilitate the development of novel diagnostic and therapeutic approaches. In this review, the biological characterization of CAFs and the mechanisms of their effects on TME remodeling are summarized. Furthermore, we also highlight currently available therapeutic strategies targeting CAF in the context of optimizing the success of immunotherapies and briefly discuss possible future perspectives and challenges related to CAF studies.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurosurgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yang Wang
- Department of Neurosurgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China; Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing 100070, China; Beijing Laboratory of Biomedical Materials, Beijing 100070, China.
| |
Collapse
|
40
|
Secretome of Stromal Cancer-Associated Fibroblasts (CAFs): Relevance in Cancer. Cells 2023; 12:cells12040628. [PMID: 36831295 PMCID: PMC9953839 DOI: 10.3390/cells12040628] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The cancer secretome reflects the assortment of proteins released by cancer cells. Investigating cell secretomes not only provides a deeper knowledge of the healthy and transformed state but also helps in the discovery of novel biomarkers. Secretomes of cancer cells have been studied in the past, however, the secretome contribution of stromal cells needs to be studied. Cancer-associated fibroblasts (CAFs) are one of the predominantly present cell populations in the tumor microenvironment (TME). CAFs play key role in functions associated with matrix deposition and remodeling, reciprocal exchange of nutrients, and molecular interactions and signaling with neighboring cells in the TME. Investigating CAFs secretomes or CAFs-secreted factors would help in identifying novel CAF-specific biomarkers, unique druggable targets, and an improved understanding for personalized cancer diagnosis and prognosis. In this review, we have tried to include all studies available in PubMed with the keywords "CAFs Secretome". We aim to provide a comprehensive summary of the studies investigating role of the CAF secretome on cancer development, progression, and therapeutic outcome. However, challenges associated with this process have also been addressed in the later sections. We have highlighted the functions and clinical relevance of secretome analysis in stromal CAF-rich cancer types. This review specifically discusses the secretome of stromal CAFs in cancers. A deeper understanding of the components of the CAF secretome and their interactions with cancer cells will help in the identification of personalized biomarkers and a more precise treatment plan.
Collapse
|
41
|
Therapeutic Targeting of Cancer-Associated Fibroblasts in the Non-Small Cell Lung Cancer Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15020335. [PMID: 36672284 PMCID: PMC9856659 DOI: 10.3390/cancers15020335] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Lung cancer is the most frequently diagnosed cancer and the leading cause of cancer death worldwide. The most common lung cancer is non-small cell lung cancer (NSCLC), with an overall 5-year survival rate of around 20% because NSCLC is a metastatic disease. A better understanding of the mechanism underlying lung cancer metastasis is therefore urgently needed. The tumor microenvironment involves different types of stromal cells and functions as key components in the progression of NSCLC. Through epithelial-mesenchymal transition (EMT), in which epithelial cells lose their polarity and acquire mesenchymal potential, cancer cells acquire metastatic abilities, as well as cancer stem-cell-like potential. We previously reported that cancer-associated fibroblasts (CAFs) interact with lung cancer cells to allow for the acquisition of malignancy and treatment resistance by paracrine loops via EMT signals in the tumor microenvironment. Furthermore, CAFs regulate the cytotoxic activity of immune cells via various cytokines and chemokines, creating a microenvironment of immune tolerance. Regulation of CAFs can therefore affect immune responses. Recent research has shown several roles of CAFs in NSCLC tumorigenesis, owing to their heterogeneity, so molecular markers of CAFs should be elucidated to better classify tumor-promoting subtypes and facilitate the establishment of CAF-specific targeted therapies. CAF-targeted cancer treatments may suppress EMT and regulate the niche of cancer stem cells and the immunosuppressive network and thus may prove useful for NSCLC treatment through multiple mechanisms.
Collapse
|
42
|
Fang Z, Meng Q, Xu J, Wang W, Zhang B, Liu J, Liang C, Hua J, Zhao Y, Yu X, Shi S. Signaling pathways in cancer-associated fibroblasts: recent advances and future perspectives. Cancer Commun (Lond) 2023; 43:3-41. [PMID: 36424360 PMCID: PMC9859735 DOI: 10.1002/cac2.12392] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/20/2022] [Accepted: 11/04/2022] [Indexed: 11/26/2022] Open
Abstract
As a critical component of the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) play important roles in cancer initiation and progression. Well-known signaling pathways, including the transforming growth factor-β (TGF-β), Hedgehog (Hh), Notch, Wnt, Hippo, nuclear factor kappa-B (NF-κB), Janus kinase (JAK)/signal transducer and activator of transcription (STAT), mitogen-activated protein kinase (MAPK), and phosphoinositide 3-kinase (PI3K)/AKT pathways, as well as transcription factors, including hypoxia-inducible factor (HIF), heat shock transcription factor 1 (HSF1), P53, Snail, and Twist, constitute complex regulatory networks in the TME to modulate the formation, activation, heterogeneity, metabolic characteristics and malignant phenotype of CAFs. Activated CAFs remodel the TME and influence the malignant biological processes of cancer cells by altering the transcriptional and secretory characteristics, and this modulation partially depends on the regulation of signaling cascades. The results of preclinical and clinical trials indicated that therapies targeting signaling pathways in CAFs demonstrated promising efficacy but were also accompanied by some failures (e.g., NCT01130142 and NCT01064622). Hence, a comprehensive understanding of the signaling cascades in CAFs might help us better understand the roles of CAFs and the TME in cancer progression and may facilitate the development of more efficient and safer stroma-targeted cancer therapies. Here, we review recent advances in studies of signaling pathways in CAFs and briefly discuss some future perspectives on CAF research.
Collapse
Affiliation(s)
- Zengli Fang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Qingcai Meng
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jin Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Wei Wang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Bo Zhang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jiang Liu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Chen Liang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jie Hua
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Yingjun Zhao
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Si Shi
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| |
Collapse
|
43
|
Zulaziz N, Chai SJ, Lim KP. The origins, roles and therapies of cancer associated fibroblast in liver cancer. Front Oncol 2023; 13:1151373. [PMID: 37035187 PMCID: PMC10076538 DOI: 10.3389/fonc.2023.1151373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer. It is often preceded by chronic inflammation such as liver fibrosis and cirrhosis. Different cell types are believed to give rise to liver-specific cancer associated fibroblast (CAF), these include resident fibroblast, hepatic stellate cell, liver cancer cell, hepatic sinusoidal endothelial cell and mesenchymal stromal cell. The abundance of fibroblasts has contributed to the cancer progression, immune modulation and treatment resistance in HCC. In this review, we discussed the origins, subtypes and roles of cancer associated fibroblasts in HCC. Their specific roles in shaping the tumor microenvironment, facilitating cancer growth, and modulating different immune cell types to confer a permissive environment for cancer growth. CAF is now an attractive therapeutic target for cancer treatment, however specific therapeutic development in HCC is still lacking. Hence, we have included preclinical and clinical development of CAF-specific interventions for other cancer types in this review. However, most CAF-specific therapies have resulted in disappointing clinical outcomes, likely due to the difficulties in differentiating CAF from normal fibroblast. A thorough understanding of the characteristics and functionalities of CAF is warranted to further improve the therapeutic efficacy of anti-CAF therapies.
Collapse
|
44
|
Zhang J, Hu Z, Horta CA, Yang J. Regulation of epithelial-mesenchymal transition by tumor microenvironmental signals and its implication in cancer therapeutics. Semin Cancer Biol 2023; 88:46-66. [PMID: 36521737 DOI: 10.1016/j.semcancer.2022.12.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been implicated in various aspects of tumor development, including tumor invasion and metastasis, cancer stemness, and therapy resistance. Diverse stroma cell types along with biochemical and biophysical factors in the tumor microenvironment impinge on the EMT program to impact tumor progression. Here we provide an in-depth review of various tumor microenvironmental signals that regulate EMT in cancer. We discuss the molecular mechanisms underlying the role of EMT in therapy resistance and highlight new therapeutic approaches targeting the tumor microenvironment to impact EMT and tumor progression.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Zhimin Hu
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Calista A Horta
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
45
|
Niu ZS, Wang WH, Niu XJ. Recent progress in molecular mechanisms of postoperative recurrence and metastasis of hepatocellular carcinoma. World J Gastroenterol 2022; 28:6433-6477. [PMID: 36569275 PMCID: PMC9782839 DOI: 10.3748/wjg.v28.i46.6433] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/31/2022] [Accepted: 11/21/2022] [Indexed: 12/08/2022] Open
Abstract
Hepatectomy is currently considered the most effective option for treating patients with early and intermediate hepatocellular carcinoma (HCC). Unfortunately, the postoperative prognosis of patients with HCC remains unsatisfactory, predominantly because of high postoperative metastasis and recurrence rates. Therefore, research on the molecular mechanisms of postoperative HCC metastasis and recurrence will help develop effective intervention measures to prevent or delay HCC metastasis and recurrence and to improve the long-term survival of HCC patients. Herein, we review the latest research progress on the molecular mechanisms underlying postoperative HCC metastasis and recurrence to lay a foundation for improving the understanding of HCC metastasis and recurrence and for developing more precise prevention and intervention strategies.
Collapse
Affiliation(s)
- Zhao-Shan Niu
- Laboratory of Micromorphology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Wen-Hong Wang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Xiao-Jun Niu
- Department of Internal Medicine, Qingdao Shibei District People's Hospital, Qingdao 266033, Shandong Province, China
| |
Collapse
|
46
|
Wu Y, Clark KC, Nguyen EV, Niranjan B, Horvath LG, Taylor RA, Daly RJ. Proteomic characterisation of prostate cancer intercellular communication reveals cell type-selective signalling and TMSB4X-dependent fibroblast reprogramming. Cell Oncol (Dordr) 2022; 45:1311-1328. [PMID: 36169805 DOI: 10.1007/s13402-022-00719-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In prostate cancer, the tumour microenvironment (TME) represents an important regulator of disease progression and response to treatment. In the TME, cancer-associated fibroblasts (CAFs) play a key role in tumour progression, however the mechanisms underpinning fibroblast-cancer cell interactions are incompletely resolved. Here, we address this by applying cell type-specific labelling with amino acid precursors (CTAP) and mass spectrometry (MS)-based (phospho)proteomics to prostate cancer for the first time. METHODS Reciprocal interactions between PC3 prostate cancer cells co-cultured with WPMY-1 prostatic fibroblasts were characterised using CTAP-MS. Signalling network changes were determined using Metascape and Enrichr and visualised using Cytoscape. Thymosin β4 (TMSB4X) overexpression was achieved via retroviral transduction and assayed by ELISA. Cell motility was determined using Transwell and random cell migration assays and expression of CAF markers by indirect immunofluorescence. RESULTS WPMY-1 cells co-cultured with PC3s demonstrated a CAF-like phenotype, characterised by enhanced PDGFRB expression and alterations in signalling pathways regulating epithelial-mesenchymal transition, cytoskeletal organisation and cell polarisation. In contrast, co-cultured PC3 cells exhibited more modest network changes, with alterations in mTORC1 signalling and regulation of the actin cytoskeleton. The expression of the actin binding protein TMSB4X was significantly decreased in co-cultured WPMY-1 fibroblasts, and overexpression of TMSB4X in fibroblasts decreased migration of co-cultured PC3 cells, reduced fibroblast motility, and protected the fibroblasts from being educated to a CAF-like phenotype by prostate cancer cells. CONCLUSIONS This study highlights the potential of CTAP-MS to characterise intercellular communication within the prostate TME and identify regulators of cellular crosstalk such as TMSB4X.
Collapse
Affiliation(s)
- Yunjian Wu
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Kimberley C Clark
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Elizabeth V Nguyen
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Birunthi Niranjan
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Lisa G Horvath
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- University of Sydney, Camperdown, NSW, 2006, Australia
- Chris O'Brien Lifehouse, Camperdown, NSW, 2050, Australia
| | - Renea A Taylor
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Physiology, Monash University, Clayton, VIC, 3800, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, 3800, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
47
|
Yang L, Qiao P, Zhang J, Chen X, Hu A, Huang S. Crosstalk between ROCK1 and PYROXD1 regulates CAFs activation and promotes laryngeal squamous cell carcinoma metastasis. Discov Oncol 2022; 13:120. [PMID: 36334145 PMCID: PMC9637080 DOI: 10.1007/s12672-022-00578-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/15/2022] [Indexed: 11/06/2022] Open
Abstract
We previously found that the Rho-associated kinase 1 (ROCK1) activated Cancer-associated fibroblasts (CAFs) to promote LSCC metastasis. Accumulating evidence indicates that pyridine nucleotide-disulfide oxidoreductase domain 1 (PYROXD1) is an oncogene; however, the crosstalk between ROCK1 and PYROXD1 in LSCC metastasis remains largely unknown. Here, we found that ROCK1 could target PYROXD1. The knockdown of ROCK1 expression reduces the expression of PYROXD1, while the knockdown of PYROXD1 expression did not alter the expression of ROCK1 indicating that ROCK1 is upstream of PYROXD1. Further, LSCC cells cocultured with PYROXD1 knocked-down CAFs exhibited lower proliferation, migration, invasion and metastasis abilities. Conversely, LSCC cells cocultured with PYROXD1-overexpressing CAFs showed opposite results. In conclusion, the crosstalk between ROCK1 and PYROXD1 regulated CAFs activation and promoted LSCC metastasis.
Collapse
Affiliation(s)
- Liyun Yang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Peipei Qiao
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Jianwei Zhang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Xiaoping Chen
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - An Hu
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - Shuixian Huang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
48
|
Huang L, Wang L, Shi Y, Zhao Y, Xu C, Zhang J, Hu W. Brain metastasis from gastric adenocarcinoma: A large comprehensive population-based cohort study on risk factors and prognosis. Front Oncol 2022; 12:897681. [PMID: 36338733 PMCID: PMC9635449 DOI: 10.3389/fonc.2022.897681] [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: 03/16/2022] [Accepted: 09/14/2022] [Indexed: 01/19/2023] Open
Abstract
Aims Although brain metastasis from gastric adenocarcinoma (GaC) is rare, it may significantly affect survival and quality of life. The aim of this large, comprehensive, population-based cohort investigation was to investigate factors that were associated with brain metastasis from GaC and to explore the prognostic factors and time-dependent cumulative mortalities among cases with GaC and brain involvement. Methods Population-based information on cases with GaC diagnosed from 2010 to 2016 was obtained from a large-scale database. Factors that were associated with brain metastasis were investigated utilizing multivariable logistic regression. Time-dependent tumor-specific mortalities of cases with GaC and brain involvement were then computed utilizing the cumulative incidence functions (CIFs), and mortalities were compared between subgroups utilizing Gray's test. Factors that were associated with death were further evaluated utilizing multivariable Fine-Gray subdistribution hazard regression. Results Together, 28,736 eligible cases were included, which comprised 231 (1%) cases with brain metastasis and 10,801 (38%) with metastasis to other sites, encompassing a follow-up of 39,168 person-years. Brain metastasis occurred more often among younger patients (within overall cancers), in cases with stomach cardia tumors, within cases with signet-ring cell carcinoma (within overall cancers), and within cases with positive lymph nodes (within overall tumors); it was less often detected among black people. Brain involvement was associated with more lung and bone metastases. The median survival time of cases having brain metastasis was only 3 months; the 6- and 12-month tumor-specific cumulative mortalities were 57% and 71%, respectively. Among cases with GaC and brain metastasis, those with gastric cardia cancers (when receiving radiotherapy), those undergoing resection, and those receiving chemotherapy had lower mortality risks, while younger patients (when receiving chemotherapy or radiotherapy) and people with positive lymph nodes (when receiving radiotherapy) had higher death hazards. Conclusion Among patients with GaC, brain metastasis was correlated with several clinical and pathological variables, including ethnicity, age, cancer histology, location, lymph node involvement, and metastases to other sites. Cases having brain metastasis had poor survival that was correlated with age, cancer location, lymph node metastasis, and management. These findings offer vital clues for individualized patient care and future mechanistic explorations.
Collapse
Affiliation(s)
- Lei Huang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital (MCARJH), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wang
- Medical Center on Aging of Ruijin Hospital (MCARJH), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yajie Zhao
- Medical Center on Aging of Ruijin Hospital (MCARJH), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenying Xu
- Medical Center on Aging of Ruijin Hospital (MCARJH), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, China
| | - Weiguo Hu
- Medical Center on Aging of Ruijin Hospital (MCARJH), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
49
|
Liu C, Wang M, Zhang H, Li C, Zhang T, Liu H, Zhu S, Chen J. Tumor microenvironment and immunotherapy of oral cancer. Eur J Med Res 2022; 27:198. [PMID: 36209263 PMCID: PMC9547678 DOI: 10.1186/s40001-022-00835-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
Oral cancer is one of the most common malignant tumors of the head and neck, not only affects the appearance, but also affects eating and even endangers life. The clinical treatments of oral cancer mainly include surgery, radiotherapy, and chemotherapy. However, unsatisfactory therapeutic effect and toxic side effects are still the main problems in clinical treatment. Tumor microenvironment (TME) is not only closely related to the occurrence, growth, and metastasis of tumor but also works in the diagnosis, prevention, and treatment of tumor and prognosis. Future studies should continue to investigate the relationship of TME and oral cancer therapy. This purpose of this review was to analyze the characteristics of oral cancer microenvironment, summarize the traditional oral cancer therapy and immunotherapy strategies, and finally prospect the development prospects of oral cancer immunotherapy. Immunotherapy targeting tumor microenvironment is expected to provide a new strategy for clinical treatment of oral cancer.
Collapse
Affiliation(s)
- Chang Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Min Wang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Haiyang Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Chunyan Li
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Tianshou Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Hong Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Song Zhu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China.
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China.
| |
Collapse
|
50
|
Rimal R, Desai P, Daware R, Hosseinnejad A, Prakash J, Lammers T, Singh S. Cancer-associated fibroblasts: Origin, function, imaging, and therapeutic targeting. Adv Drug Deliv Rev 2022; 189:114504. [PMID: 35998825 DOI: 10.1016/j.addr.2022.114504] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/10/2022] [Accepted: 08/17/2022] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment (TME) is emerging as one of the primary barriers in cancer therapy. Cancer-associated fibroblasts (CAF) are a common inhabitant of the TME in several tumor types and play a critical role in tumor progression and drug resistance via different mechanisms such as desmoplasia, angiogenesis, immune modulation, and cancer metabolism. Due to their abundance and significance in pro-tumorigenic mechanisms, CAF are gaining attention as a diagnostic target as well as to improve the efficacy of cancer therapy by their modulation. In this review, we highlight existing imaging techniques that are used for the visualization of CAF and CAF-induced fibrosis and provide an overview of compounds that are known to modulate CAF activity. Subsequently, we also discuss CAF-targeted and CAF-modulating nanocarriers. Finally, our review addresses ongoing challenges and provides a glimpse into the prospects that can spearhead the transition of CAF-targeted therapies from opportunity to reality.
Collapse
Affiliation(s)
- Rahul Rimal
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Prachi Desai
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forkenbeckstrasse 50, 52074 Aachen, Germany
| | - Rasika Daware
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Aisa Hosseinnejad
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forkenbeckstrasse 50, 52074 Aachen, Germany
| | - Jai Prakash
- Department of Advanced Organ Bioengineering and Therapeutics, Section: Engineered Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Smriti Singh
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120 Heidelberg, Germany.
| |
Collapse
|