1
|
Lu G, Liu H, Wang H, Luo S, Du M, Christiani DC, Wei Q. Genetic variants of FER and SULF1 in the fibroblast-related genes are associated with non-small-cell lung cancer survival. Int J Cancer 2025; 156:2107-2117. [PMID: 39707607 PMCID: PMC11971011 DOI: 10.1002/ijc.35305] [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: 07/21/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Fibroblasts are important components in the tumor microenvironment and can affect tumor progression and metastasis. However, the roles of genetic variants of the fibroblast-related genes (FRGs) in the prognosis of non-small-cell lung cancer (NSCLC) patients have not been reported. Therefore, we investigated the associations between 26,544 single nucleotide polymorphisms (SNPs) in 291 FRGs and survival of NSCLC patients from the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. In Cox regression multivariable analysis, we found that 661 SNPs were associated with NSCLC overall survival (OS). Then we validated these SNPs in another independent replication dataset of 984 patients from the Harvard Lung Cancer Susceptibility (HLCS) Study. Finally, we identified two independent SNPs (i.e., FER rs7716388 A>G and SULF1 rs11785839 G>C) that remained significantly associated with NSCLC survival with hazards ratios (HRs) of 0.87 (95% confidence interval [CI] = 0.77-0.98, p = 0.018) and 0.88 (95% CI = 0.79-0.99, p = 0.033), respectively. Combined analysis for these two SNPs showed that the number of protective alleles was associated with better OS and disease-specific survival. Expression quantitative trait loci analysis indicated that the FER rs7716388 G allele was associated with the up-regulation of FER mRNA expression levels in lung tissue. Our results indicated that these two functional SNPs in the FRGs may be prognostic biomarkers for the prognosis of NSCLC patients, and the possible mechanism may be through modulating the expression of their corresponding genes.
Collapse
Affiliation(s)
- Guojun Lu
- Department of Respiratory Medicine, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Huilin Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Respiratory Oncology, Guangxi Cancer Hospital, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mulong Du
- Departments of Environmental Health and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115 USA
| | - David C. Christiani
- Departments of Environmental Health and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115 USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University Medical Center, Durham, Durham, NC 27710, USA
- Duke Global Health Institute, Duke University Medical Center, Durham, Durham, NC 27710, USA
| |
Collapse
|
2
|
Feng Y, Jiang Y, Yang L, Lu D, Li N, Zhang Q, Yang H, Qin H, Zhang J, Gou X, Jiang F. Targeting CAFs and extracellular matrix (ECM) in lung cancer: Potential of adjuvants and nanoparticles. Bioorg Chem 2025; 162:108586. [PMID: 40398184 DOI: 10.1016/j.bioorg.2025.108586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/09/2025] [Accepted: 05/11/2025] [Indexed: 05/23/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are prominent components of the lung tumor stroma and are known to foster tumor growth, invasion, and metastasis through extracellular matrix (ECM) and tumor stroma remodeling. The interactions of CAFs with cancer cells and other stromal components contribute significantly to the aggressive nature of lung cancer and pose challenges to conventional treatment approaches. Simultaneously, the ECM, which contains numerous proteins and other molecules surrounding cancer cells, serves as more than just a structural scaffold. In lung cancer, alterations in ECM composition and organization not only promote tumor cell proliferation and survival but also impact drug penetration, immune cell infiltration, and therapeutic resistance. Targeting the intricate interplay between CAFs and the dynamic ECM in lung cancer represents a crucial frontier in oncology research. This review aims to delve deeply into the pivotal roles of CAFs and the ECM in the tumorigenesis and progression of lung cancer. Then, the potential of utilizing adjuvants, phytochemicals, and nanoparticles to modulate the functions of CAFs and remodel the ECM in the lung tumor will be reviewed.
Collapse
Affiliation(s)
- Yuan Feng
- Doctoral student of Guangxi University of Chinese Medicine in grade 2022, Nanning, Guangxi 530200, China
| | - Ying Jiang
- Department of Neurology, Ruikang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi 530011, China
| | - Lin Yang
- Master student of Guangxi University of Chinese Medicine in 2022, Nanning, Guangxi 530200, China
| | - Danni Lu
- Master student of Guangxi University of Chinese Medicine in 2022, Nanning, Guangxi 530200, China
| | - Ning Li
- Master student of Guangxi University of Chinese Medicine in 2023, Nanning, Guangxi 530200, China
| | - Qun Zhang
- Master student of Guangxi University of Chinese Medicine in 2023, Nanning, Guangxi 530200, China
| | - Haiyan Yang
- Master student of Guangxi University of Chinese Medicine in 2023, Nanning, Guangxi 530200, China
| | - Huiyuan Qin
- Master student of Guangxi University of Chinese Medicine in 2024, Nanning, Guangxi 530200, China
| | - Jiaxin Zhang
- Master student of Guangxi University of Chinese Medicine in 2024, Nanning, Guangxi 530200, China
| | - Xinyun Gou
- Master student of Guangxi University of Chinese Medicine in 2024, Nanning, Guangxi 530200, China
| | - Feng Jiang
- Science and Technology Department of Ruikang Hospital affiliated to Guangxi University of Chinese Medicine, 530011, China.
| |
Collapse
|
3
|
Zhong H, Zhou S, Yin S, Qiu Y, Liu B, Yu H. Tumor microenvironment as niche constructed by cancer stem cells: Breaking the ecosystem to combat cancer. J Adv Res 2025; 71:279-296. [PMID: 38866179 DOI: 10.1016/j.jare.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a distinct subpopulation of cancer cells with the capacity to constantly self-renew and differentiate, and they are the main driver in the progression of cancer resistance and relapse. The tumor microenvironment (TME) constructed by CSCs is the "soil" adapted to tumor growth, helping CSCs evade immune killing, enhance their chemical resistance, and promote cancer progression. AIM OF REVIEW We aim to elaborate the tight connection between CSCs and immunosuppressive components of the TME. We attempt to summarize and provide a therapeutic strategy to eradicate CSCs based on the destruction of the tumor ecological niche. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight that CSCs recruit and transform normal cells to construct the TME, which further provides ecological niche support for CSCs. Second, we describe the main characteristics of the immunosuppressive components of the TME, targeting strategies and summarize the progress of corresponding drugs in clinical trials. Third, we explore the multilevel insights of the TME to serve as an ecological niche for CSCs.
Collapse
Affiliation(s)
- Hao Zhong
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
4
|
FENG LIWEN, CHEN YUTING, JIN WENYI. Research progress on cancer-associated fibroblasts in osteosarcoma. Oncol Res 2025; 33:1091-1103. [PMID: 40296919 PMCID: PMC12033999 DOI: 10.32604/or.2024.054207] [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: 05/21/2024] [Accepted: 08/26/2024] [Indexed: 04/30/2025] Open
Abstract
Osteosarcoma (OS) is a prevalent primary bone malignancy with limited treatment options. Therefore, it is imperative to investigate and understand the mechanisms underlying OS pathogenesis. Cancer-associated fibroblasts (CAFs) are markedly abundant in tumor stromal cells and are essentially involved in the modulation of tumor occurrence and development. In recent years, CAFs have become a hotspot as researchers aim to elucidate CAF mechanisms that regulate tumor progression. However, most studies on CAFs are limited to a few common cancers, and their association with OS remains elusive. This review describes the role and current knowledge of CAFs in OS, focusing on their potential cellular origin, classification, and diverse functionality. It was found that CAFs influenced OS tumor cell signaling, proliferation, invasion, metastasis, epithelial-mesenchymal transition, stemness maintenance, angiogenesis, and the ability to modify immune system components. Furthermore, findings on other common cancers indicated that effective therapeutic strategies included the manipulation of CAF activation, targeting CAF-derived components, and depletion of CAFs by biomarkers. This review provides new insights and a theoretical basis for OS research.
Collapse
Affiliation(s)
- LIWEN FENG
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - YUTING CHEN
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - WENYI JIN
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430022, China
| |
Collapse
|
5
|
Whitman MA, Mantri M, Spanos E, Estroff LA, De Vlaminck I, Fischbach C. Bone mineral density affects tumor growth by shaping microenvironmental heterogeneity. Biomaterials 2025; 315:122916. [PMID: 39490060 DOI: 10.1016/j.biomaterials.2024.122916] [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: 07/19/2024] [Revised: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Breast cancer bone metastasis is a major cause of mortality in patients with advanced breast cancer. Although decreased mineral density is a known risk factor for bone metastasis, the underlying mechanisms remain poorly understood because studying the isolated effect of bone mineral density on tumor heterogeneity is challenging with conventional approaches. Moreover, mineralized biomaterials are commonly utilized for clinical bone defect repair, but how mineralized biomaterials affect the foreign body response and wound healing is unclear. Here, we investigate how bone mineral affects tumor growth and microenvironmental complexity in vivo by combining single-cell RNA-sequencing with mineral-containing or mineral-free decellularized bone matrices. We discover that the absence of bone mineral significantly influences fibroblast and immune cell heterogeneity, promoting phenotypes that increase tumor growth and alter the response to injury or disease. Importantly, we observe that the stromal response to bone mineral content depends on the murine tumor model used. While lack of bone mineral induces tumor-promoting microenvironments in both immunocompromised and immunocompetent animals, these changes are mediated by altered fibroblast phenotype in immunocompromised mice and macrophage polarization in immunocompetent mice. Collectively, our findings suggest that bone mineral density affects tumor growth by impacting microenvironmental complexity in an organism-dependent manner.
Collapse
Affiliation(s)
- Matthew A Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Emmanuel Spanos
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14850, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14850, USA
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA.
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14850, USA.
| |
Collapse
|
6
|
Xu Y, Sun D, He J, Yao Q. Cancer-associated fibroblasts in ovarian cancer: research progress. Front Oncol 2025; 15:1504762. [PMID: 40104498 PMCID: PMC11913710 DOI: 10.3389/fonc.2025.1504762] [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: 10/01/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Ovarian cancer, known for its high invasiveness and therapeutic resistance, is one of the leading causes of death from gynecological tumors. The tumor microenvironment (TME) plays a crucial role in the development of ovarian cancer, with cancer-associated fibroblasts (CAFs) being a key non-tumor cell component. They significantly affect the prognosis of ovarian cancer by promoting tumor cell proliferation, invasion, metastasis, immune evasion, and drug resistance. The heterogeneity of CAFs provides a new perspective for targeted therapy in ovarian cancer. This review comprehensively analyzes the mechanisms of action, heterogeneity characteristics, and role in the immune microenvironment of CAFs in ovarian cancer, and discusses targeted therapy strategies for CAFs, aiming to provide new theoretical basis and treatment directions for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yuance Xu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Danting Sun
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junqi He
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qin Yao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Que Y, Ding T, Wang H, Xu S, He P, Shen Q, Cao K, Luo Y, Hu Y. Prognostic and Therapeutic Significance of Cancer-Associated Fibroblasts Genes in Osteosarcoma Based on Bulk and Single-Cell RNA Sequencing Data. J Cell Mol Med 2025; 29:e70424. [PMID: 40045162 PMCID: PMC11882394 DOI: 10.1111/jcmm.70424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 11/25/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Osteosarcoma (OS) is the most frequent primary solid malignancy of bone, whose course is usually dismal without efficient treatments. The aim of the study was to discover novel risk models to more accurately predict and improve the prognosis of patients with osteosarcoma. The single-cell RNA sequencing (scRNA-seq) data was obtained from the GEO database. Bulk RNA-seq data and microarray data of OS were obtained from the TARGET and GEO databases respectively. A clustering tree was plotted to classify all cells into different clusters. The "cellchat" R package was used to establish and visualise cell-cell interaction networks. Then Univariate COX regression analysis was used to determine the prognostic CAF-related genes, followed by the Lasso-Cox regression analysis to build a risk on the prognostic CAF-related genes. Finally, from multiple perspectives, the signature was validated as an accurate and dependable tool in predicting the prognosis and guiding treatment therapies in OS patients. From the single-cell dataset, six OS patients and 46,544 cells were enrolled. All cells were classified into 22 clusters, and the clusters were annotated to 14 types of cells. Subsequently, CAFs were observed as a vital TME components. In cell-cell interaction networks in OS cells, CAFs had a profound impact as four roles. Via the Univariate COX regression analysis, 14 CAF-related genes were screened out. By the Lasso-Cox regression analyses, 11 key CAF-related genes were obtained, based on which an 11-gene signature that could predict the prognosis of osteosarcoma patients was constructed. According to the median of risk scores, all patients were grouped in to the high- and low-risk group, and their overall survival, activated pathways, immune cell infiltrations, and drug sensitivity were significantly differential, which may have important implications for the clinical treatment of patients with osteosarcoma. Our study, a systematic analysis of gene and regulatory genes, has proven that CAF-related genes had excellent diagnostic and prognostic capabilities in OS, and it may reshape the TME in OS. The novel CAF-related risk signature can effectively predict the prognosis of OS and provide new strategies for cancer treatment.
Collapse
Affiliation(s)
- Yukang Que
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| | - Tianming Ding
- Department of OrthopedicsYangzhou East HospitalYangzhouJiangsuChina
| | - Huming Wang
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| | - Shenglin Xu
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| | - Peng He
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| | - Qiling Shen
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| | - Kun Cao
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| | - Yang Luo
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| | - Yong Hu
- Department of OrthopedicsThe First Affiliated Hospital Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
8
|
Zhang Z, Tang Y, Luo D, Qiu J, Chen L. Advances in nanotechnology for targeting cancer-associated fibroblasts: A review of multi-strategy drug delivery and preclinical insights. APL Bioeng 2025; 9:011502. [PMID: 40094065 PMCID: PMC11910205 DOI: 10.1063/5.0244706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment by promoting tumor growth, immune evasion, and metastasis. Recently, drug delivery systems targeting CAFs have emerged as a promising long-term and effective approach to cancer treatment. Advances in nanotechnology, in particular, have led to the development of nanomedicine delivery systems designed specifically to target CAFs, offering new possibilities for precise and personalized cancer therapies. This article reviews recent progress in drug delivery using nanocarriers that target CAFs. Additionally, we explore the potential of combining multiple therapies, such as chemotherapy and immunotherapy, with nanocarriers to enhance efficacy and overcome drug resistance. Although many preclinical studies show promise, the clinical application of nanomedicine still faces considerable challenges, especially in terms of drug penetration and large-scale production. Therefore, this review aims to provide a fresh perspective on CAF-targeted drug delivery systems and highlight potential future research directions and clinical applications.
Collapse
|
9
|
Sun L, Zhang HB, Jiang HC, Li W, Li MK, Yang XY, Cai YY, Xue KK, Gou YS, Liu XY, Liang Q, Zuo LG, Hu JG, Qian F. LMO7 drives profibrotic fibroblast polarization and pulmonary fibrosis in mice through TGF-β signalling. Acta Pharmacol Sin 2025:10.1038/s41401-025-01488-9. [PMID: 40000880 DOI: 10.1038/s41401-025-01488-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lethal disease. Profibrotic fibroblast polarization during wound healing is one of the main causes of IPF, and the molecular mechanisms involved have yet to be fully determined. LIM domain-only protein 7 (LMO7), which acts as an E3 ubiquitin ligase, is highly expressed in the lung, brain and heart and plays important roles in embryonic development, cancer progression, inflammatory bowel disease and Dreifuss muscular dystrophy (EDMD). In this study, we investigated the role of LMO7 in pulmonary fibrosis. Bleomycin (BLM)-induced lung fibrosis was established in mice. For AAV-mediated gene therapy, AAV-Lmo7 shRNA (AAV-Lmo7 shRNA) was intratracheally administered 6 days before BLM injection. Through transcriptome analysis, we found that the expression of LMO7 was significantly upregulated in the fibroblasts of IPF patients and BLM-induced mice. Knockdown of LMO7 impaired the profibrotic phenotype of fibroblasts in BLM-treated mice and in primary lung fibroblasts stimulated with TGF-β in vitro. We observed that LMO7 binds to SMAD7, mediating its degradation by polyubiquitination of lysine 70 and increasing the stability of TGF-β receptor 1 (TGFβR1). Finally, intratracheal administration of adeno-associated virus (AAV)-mediated Lmo7 shRNA significantly ameliorated the progression of BLM-induced lung fibrosis. Our results suggest that LMO7 is a promising target for blocking profibrotic fibroblast polarization for the treatment of fibrotic lung disease. A model for the role of LMO7 in TGF-β/SMAD signaling during pulmonary fibrosis. During pulmonary fibrosis, ubiquitin E3 ligase LMO7 is up-regulated, and binds with. SMAD7. LMO7 mediates the ubiquitination of SMAD7 on Lysine 70, leading to its degradation, and further enhances the stability of transforming growth factor-beta receptor 1 (TGFβR1).
Collapse
Affiliation(s)
- Lei Sun
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Hai-Bo Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hong-Chao Jiang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wen Li
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Meng-Kai Li
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xin-Yi Yang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuan-Yuan Cai
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ke-Ke Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yu-Sen Gou
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xin-Yue Liu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qing Liang
- Department of Pharmacy, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Lu-Gen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
| | - Jian-Guo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China.
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China.
| | - Feng Qian
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China.
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
10
|
Kumar BV, Sachan R, Garad P, Srivastava N, Saraf SA, Meher N. Dual Targeting of Prostate-Specific Membrane Antigen and Fibroblast Activation Protein: Bridging Prostate Cancer Theranostics with Precision. ACS APPLIED BIO MATERIALS 2025; 8:962-979. [PMID: 39895503 DOI: 10.1021/acsabm.4c01914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Targeting Prostate Specific Membrane Antigen (PSMA) has proven highly useful and beneficial for prostate cancer (PCa) theranostics. However, patients with advanced metastatic castration-resistant prostate cancer (mCRPC) lack optimal PSMA expression resulting in poor specificity. To address this limitation, combination targeting is gaining popularity by synergistically boosting the theranostic efficacy. Herein, we thoroughly reviewed the most recent development of drug formulation for PCa theranostics by targeting both PSMA and Fibroblast Activation Protein (FAP). FAP is known to overexpress in cancer-associated fibroblasts (CAFs) within the tumor microenvironment (TME). It has been extensively studied as an effective target for the identification and treatment of a variety of cancer phenotypes. Along with the advantages and current updates on combination targeting of PSMA and FAP, this Review thoroughly discussed the expression patterns of PSMA and FAP in various cancer phenotypes, as well as their role in tumor growth, invasion, and metastasis, which is of great interest in the design and development of prostate cancer theranostics.
Collapse
Affiliation(s)
- Boga Vijay Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Riya Sachan
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Prajakta Garad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Shubhini A Saraf
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Niranjan Meher
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, Uttar Pradesh 226002, India
| |
Collapse
|
11
|
Wang Y, Zhang Z, Li X, Lu X, Zhuansun X, Li Q, Zhang J, Xu X, Liu X, Wei Y, Hua F, Wu R, Chen Z. Colorectal carcinoma organoid and cancer-associated fibroblasts co-culture system for drug evaluation. IN VITRO MODELS 2025; 4:31-44. [PMID: 40160212 PMCID: PMC11950461 DOI: 10.1007/s44164-025-00084-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 04/02/2025]
Abstract
Patient-derived organoids (PDO) have the potential to be used as preclinical cancer models for testing anti-cancer drug efficiency. Cancer-associated fibroblasts (CAFs), which have been closely linked with colorectal carcinoma (CRC) progression and drug resistance, however, are generally not included (or gradually lost during culture) in the PDO models, leading to a major limitation in this cancer model. In this study, we established a new in vitro model with CRC organoids and co-cultured with CAFs and compared it with the organoid-only model. Through testing with anti-cancer drug, we demonstrated a significant difference in drug sensitivity between the two models, and the co-culture model showed higher drug resistance. RNA and whole exome sequencing were performed to reveal gene expression profiles in organoids and organoids co-culture with CAFs to assess interactions between drug sensitivity and gene copy number variation. We found that the expression levels of several pathway protein genes, which are highly expressed in original surgical specimens of colorectal carcinomas, were downregulated in organoids but restored in organoids by co-culturing with CAFs. In summary, the PDO-CAF joint model for CRC can recapitulate a more biomimetic tumor microenvironment and the drug resistance lead by changes in multiple signaling pathways that we discovered; thus, it could be a suitable model for future usage in drug discovery and precision medicine research.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
| | - Zilin Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Xiaoran Li
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Xiaobing Lu
- Jiangsu Health Vocational College, Nanjing, 210029 China
| | - Xuemei Zhuansun
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Jing Zhang
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Xi Xu
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Xueqiang Liu
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Yuan Wei
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
- Jiangsu Avatarget Biotechnology Co., Ltd, Suzhou, 215163 China
| | - Feng Hua
- Department of Pharmacy, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000 China
| | - Runda Wu
- The First Affiliated Hospital of Suzhou University, Suzhou, 215006 China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, 215163 China
| |
Collapse
|
12
|
Kanzaki R, Reid S, Bolivar P, Sjölund J, Staaf J, Larsson S, Shintani Y, Pietras K. FHL2 expression by cancer-associated fibroblasts promotes metastasis and angiogenesis in lung adenocarcinoma. Int J Cancer 2025; 156:431-446. [PMID: 39244734 DOI: 10.1002/ijc.35174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024]
Abstract
Cancer-associated fibroblasts (CAFs) contribute to the progression of lung cancer. Four and a half LIM domain protein-2 (FHL2) is a component of focal adhesion structures. We analyzed the function of FHL2 expressed by CAFs in lung adenocarcinoma. Expression of FHL2 in fibroblast subtypes was investigated using database of single-cell RNA-sequencing of lung cancer tissue. The role of FHL2 in the proliferation and migration of CAFs was assessed. The effects of FHL2 knockout on the migration and invasion of human lung adenocarcinoma cells and tube formation of endothelial cells induced by CAF-conditioned medium (CM) were evaluated. The effect of FHL2 knockout in CAFs on metastasis was determined using a murine orthotopic lung cancer model. The prognostic significance of stromal FHL2 was assessed by immunohistochemistry in human adenocarcinoma specimens. FHL2 is highly expressed in myofibroblasts in cancer tissue. TGF-β1 upregulated FHL2 expression in CAFs and FHL2 knockdown attenuated CAF proliferation. FHL2 knockout reduced CAF induced migration of A110L and H23 human lung adenocarcinoma cell lines, and the induction of tube formation of endothelial cells. FHL2 knockout reduced CAF-induced metastasis of lung adenocarcinomas in an orthotopic model in vivo. The concentration of Osteopontin (OPN) in CM from CAF was downregulated by FHL2 knockout. siRNA silencing and antibody blocking of OPN reduced the pro-migratory effect of CM from CAF on lung cancer cells. In resected lung adenocarcinoma specimens, positive stromal FHL2 expression was significantly associated with higher microvascular density and worse prognosis. In conclusion, FHL2 expression by CAFs enhances the progression of lung adenocarcinoma by promoting angiogenesis and metastasis.
Collapse
Affiliation(s)
- Ryu Kanzaki
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of General Thoracic Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Steven Reid
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Paulina Bolivar
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Johan Staaf
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Sara Larsson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Gao B, Gou X, Feng C, Zhang Y, Gu H, Chai F, Wang Y, Ye Y, Hong N, Hu G, Sun B, Cheng J, Yang H. Identification of cancer-associated fibrolast subtypes and distinctive role of MFAP5 in CT-detected extramural venous invasion in gastric cancer. Transl Oncol 2025; 51:102188. [PMID: 39531783 PMCID: PMC11600027 DOI: 10.1016/j.tranon.2024.102188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/26/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Extramural venous invasion (EMVI) detected by computed tomography has been identified as an independent risk factor for distant metastasis in patients with advanced gastric cancer (GC). Cancer-associated fibroblasts (CAFs) are critical for remodeling the tumor microenvironment in GCs. Here, we report that MFAP5+ CAFs promote the formation of EMVI imaging in GC. We detected gene expression in pathological samples from 13 advanced GC patients with EMVI. Radiogenomics results showed the degree of CAFs infiltration was directly proportional to the EMVI score and EMT pathway in GC patients. Single-cell sequencing data analysis results showed that MFAP5+CAFs subtypes in GC were negatively correlated with patient prognosis and were enriched in tumor lactylation modification and EMT pathways. Immunohistochemistry results showed that the expression of MFAP5, L-lactyl and EMT markers in GC tissues was proportional to the EMVI score. CAF from gastric cancer tissue was extracted using collagenase method and co-cultured with GC cell line in vitro. After lentivirus knockdown of MFAP5 in CAFs, the levels of L-lactoyl and histone lactylation modifications were significantly reduced, and the sphere-forming and vascularization abilities of CAFs were significantly inhibited. Cell function experiments showed that MFAP5+ CAFs can affect the EMT, metastasis and invasion capabilities of GC cells. In vivo experimental results of the nude mouse in situ EMVI model suggest that MFAP5+ CAF may promote the formation of EMVI imaging features in GC by regulating lactylation modification. This innovative work may provide important new references for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Bo Gao
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Xinyi Gou
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yinli Zhang
- Department of Pathology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Huining Gu
- Department of Immunology, School of Basic Medical Sciences, Peking University and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Guohua Hu
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Boshi Sun
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China.
| | - Hao Yang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
14
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [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: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
15
|
Zhao Z, Xiong S, Gao J, Zhang Y, Guo E, Huang Y. C3 + cancer-associated fibroblasts promote tumor growth and therapeutic resistance in gastric cancer via activation of the NF-κB signaling pathway. J Transl Med 2024; 22:1130. [PMID: 39707456 DOI: 10.1186/s12967-024-05939-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) remains one of the most lethal malignancies globally, with limited therapeutic options. Cancer-associated fibroblasts (CAFs), a diverse population of stromal cells within the tumor microenvironment (TME), play a central role in tumor progression and therapeutic resistance. However, the specific markers identifying tumor-promoting CAF subsets in GC have yet to be fully characterized. METHODS Through animal studies and RNA sequencing, complement C3 (C3) emerged as a key marker linked to tumor-promoting CAF subsets. Single-cell sequencing and multiplex immunofluorescence staining confirmed that C3 expression is predominantly localized within CAFs. Independent cohort analyses demonstrated a strong association between elevated levels of C3+ CAFs and poor clinical outcomes in GC patients. To further investigate, small interfering RNA (siRNA)-mediated knockdown of C3 in CAFs was employed in vitro, with subsequent experiments, including cell migration assays, cell viability assays, and immunofluorescence, revealing significant functional impacts. RESULTS C3 secreted by CAFs promoted Epithelial-mesenchymal transition (EMT) and accelerated cancer cell migration. Patients with minimal C3+ CAF infiltration exhibited a higher probability of deriving therapeutic benefit from adjuvant treatments. Furthermore, C3+ CAFs were associated with immunosuppressive effects and an immune-evasive microenvironment marked by CD8 + T cell dysfunction. A lower prevalence of C3+ CAFs correlated with improved responsiveness to immunotherapy in GC patients. Enrichment analysis highlighted pronounced activation of the NF-κB signaling pathway in C3+ CAFs relative to their C3- counterparts, supported by elevated phosphorylation levels of IKK, IκBα, and p65 in C3+ CAFs compared to both C3- CAFs and normal fibroblasts (NFs). Silencing p65 nuclear translocation in CAFs through siRNA significantly suppressed C3 secretion. CONCLUSIONS The study suggests that NF-κB pathway-mediated CAF activation enhances C3 secretion, driving EMT, migration, chemoresistance, and immune evasion in GC progression. Targeting the NF-κB/C3 signaling axis in CAFs may offer a viable therapeutic strategy for GC management.
Collapse
Affiliation(s)
- Zhenxiong Zhao
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Si Xiong
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianpeng Gao
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yingjing Zhang
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Ergang Guo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yakai Huang
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Zhu H, Zhao C, Zhu H, Xu X, Hu C, Zhang Z. The characteristics and functional significance of disulfidptosis-related genes in head and neck squamous cell carcinoma. Discov Oncol 2024; 15:739. [PMID: 39625660 PMCID: PMC11615178 DOI: 10.1007/s12672-024-01629-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Disulfidptosis is a newfound programmed cell death (PCD) mode characterized by disulfide stress. Nevertheless, the characteristics and functional significance of disulfidptosis-related genes in head and neck squamous cell carcinoma (HNSCC) are still largely unknown. In this study, several computer-aided bioinformatic analyses were performed. The Nonnegative Matrix Factorization (NMF) method classified The Cancer Genome Atlas (TCGA) patients into two clusters according to the expression of disulfidptosis-related genes. The relative compositions of cells in the tumor microenvironment (TME), mutant landscape, lasso regression analysis, and predicted clinical outcome were performed by analyzing bulk RNA-sequencing data. Besides, single-cell sequencing data (scRNA) was analyzed by Seurat, CopyKAT, and monocle2 to reveal the expression characteristics of disulfidptosis-related genes. Moreover, the spatial distribution characteristics of each cell subgroup in the section and the functional significance of cancer-associated fibroblasts (CAFs) were elucidated by STUtility, SpaCET, and SPATA2. Here, two clusters with different expression characteristics of disulfidptosis-related genes were identified. Cluster 1 (C1) patients had a worse prognosis and a higher proportion of stromal cells but lower effector T cell infiltration than cluster 2 (C2). A novel prognostic model was established and verified in our patient cohort. Additionally, diploid and inflammatory CAFs (iCAFs) showed higher disulfidptosis-related gene expression levels. Furthermore, the CCNC and CHMP1B expressions significantly changed following CAFs differentiation. Disulfidptosis-related genes exhibited extensive and differential spatial expression on tissue sections. Collectively, our study may contribute to revealing the function of disulfidptosis, and improve the expansion of knowledge of crosstalk between cancer cells and CAFs.
Collapse
Affiliation(s)
- Haiqian Zhu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Chifeng Zhao
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Haoran Zhu
- Xi'an Jiaotong University Health Science Center, Xi'an, 710000, Shaanxi Province, China
| | - Xuhui Xu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Conglin Hu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Zhenxing Zhang
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China.
| |
Collapse
|
17
|
Mohamadi S, Mehrasa P, Mehramuz B, Kobravi S, Taghizadieh M, Salmaninejad A, Bayat M, Sadri Nahand J. The tumor microenvironment's gambit: Exosomal pawns on the board of head and neck cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189189. [PMID: 39343066 DOI: 10.1016/j.bbcan.2024.189189] [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: 07/07/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
The tumor microenvironment (TME) harbors a hidden universe of interactions that profoundly shape the behavior of head and neck cancers (HNCs). HNCs are not merely localized afflictions; they constitute a pressing global health crisis that impacts millions, frequently resulting in severe prognoses due to late-stage diagnosis and intrinsic resistance to conventional therapies. In this intricate interplay, cancer cells function as strategic players, adeptly manipulating their microenvironment to foster proliferation, evade immune detection, and withstand therapeutic interventions. Central to this dynamic play are exosomes, the enigmatic pawns of cellular communication, carrying vital messages across the board. This review elucidates the multifaceted roles of exosomes within the TME, highlighting their capacity to transmit critical signals that not only promote tumor progression but also modulate immune responses, ultimately playing a crucial role in the evolving narrative of HNC. Our insights aim to catalyze further research and exploration into exosome-targeted therapies, potentially transforming the landscape of HNC treatment and improving clinical outcomes in this formidable battle against cancer.
Collapse
Affiliation(s)
- Solmaz Mohamadi
- Faculty of Dentistry, Tabriz University of Medical Sciences, 15731 Tabriz, Iran
| | - Parisa Mehrasa
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahareh Mehramuz
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tehran Azad University, Tehran, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Salmaninejad
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, 15731 Tabriz, Iran.
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, 15731 Tabriz, Iran.
| |
Collapse
|
18
|
Li T, Huang Y, Cui S, Hong Z, Zhang X, Li Z, Chen K, Chen D. RNA methylation patterns of tumor microenvironment cells regulate prognosis and immunotherapeutic responsiveness in patients with triple-negative breast cancer. Sci Rep 2024; 14:26075. [PMID: 39478153 PMCID: PMC11525934 DOI: 10.1038/s41598-024-77941-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024] Open
Abstract
Immunotherapy research focuses on reshaping the tumor microenvironment (TME) to enhance its antitumor immune responses, with an emphasis on understanding the impact of RNA methylation in triple-negative breast cancer (TNBC) TME regulation. This study explored the influence of various RNA methyltransferases on TME cells in TNBC and their correlation with prognosis and immunotherapy response. Using non-negative matrix factorization on single-cell RNA-sequencing data, distinct TME cell clusters were identified based on the expression of 30 RNA methyltransferases. Various analyses, including pseudotime, cell communication, transcription factor regulatory network, and gene enrichment, were conducted on these clusters. The roles of RNA methyltransferase-mediated TME clusters in prognosis and immunotherapy response were determined using TNBC bulk RNA-Seq data, and the findings were validated through immunofluorescence analysis of a tissue microarray comprising 87 samples. Spatial transcriptomic analysis further revealed the distribution of TME cell clusters. Different methyltransferase-mediated cell clusters exhibited unique metabolic, immune, transcriptional, and intercellular communication patterns. Survival analysis indicated prognostic significance in specific TME cell clusters, and immunofluorescence analysis confirmed the prognostic value of m6A_WTAP + CD8T + cells. In conclusion, our study illustrated the involvement of these cell subgroups in tumor growth and antitumor immunity modulation, providing insights into the enhancement of TNBC immunotherapy.
Collapse
Affiliation(s)
- Tingjun Li
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Yiqin Huang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Shien Cui
- Breast Center of Zhongshan City People's Hospital, Zhongshan, China
| | - Zhipeng Hong
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Xinhai Zhang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Zhihao Li
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Kunqi Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Debo Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China.
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
19
|
Gao B, Hu G, Sun B, Li W, Yang H. BNIP3+ fibroblasts associated with hypoxia and inflammation predict prognosis and immunotherapy response in pancreatic ductal adenocarcinoma. J Transl Med 2024; 22:937. [PMID: 39402590 PMCID: PMC11476087 DOI: 10.1186/s12967-024-05674-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/04/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors that lacks effective treatment options. Cancer-associated fibroblasts (CAFs), an important component of the tumor microenvironment, associated with tumor progression, prognosis, and treatment response. This work aimed to explore the novel CAFs-associated target to improve treatment strategies in PDAC. METHODS The PDAC single-cell sequencing data (CRA001160, n = 35) were downloaded and integrated based on GSA databases to classify fibroblasts into fine subtypes. Functional enrichment analysis and coexpression regulatory network analysis were used to identify the functional phenotypes and biological properties of the different fibroblast subtypes. Fibroblast differentiation trajectories were constructed using pseudochronological analysis to identify initial and terminally differentiated subtypes of fibroblasts. The changes in the proportions of different fibroblast subtypes before and after PDAC immunotherapy were compared in responsive and nonresponding patients, and the relationships between fibroblast subtypes and PDAC immunotherapy responsiveness were determined based on GSA and GEO database. Using molecular biology methods to confirm the effects of BNIP3 on hypoxia and inflammation in CAFs. CAFs were co cultured with pancreatic cancer cells to detect their effects on migration and invasion of pancreatic cancer. RESULTS Single-cell data analysis divided fibroblasts into six subtypes. The differentiation trajectory suggested that BNIP3+ Fibro subtype exhibited terminal differentiation, and the expression of genes related to hypoxia and the inflammatory response increased gradually with differentiation time. The specific overexpressed genes in the BNIP3+ Fibro subtype were significantly associated with overall and disease progression-free survival in the patients with PDAC. Interestingly, the greater the proportion of the BNIP3+ Fibro subtype was, the worse the response of PDAC patients to immunotherapy, and the CRTL treatment regimen effectively reduced the proportion of the BNIP3+ Fibro subtype. After knocking out BNIP3, the hypoxia markers and inflammatory factors of CAFs were inhibited. Co-culture of CAFs with pancreatic cancer cells can increase the migration and invasion of pancreatic cancer, but this could be reversed by knocking out BNIP3. CONCLUSIONS This study revealed the BNIP3+ Fibro subtype associated with hypoxia and inflammatory responses, which was closely related to the poor prognosis of patients with PDAC, and identified signature genes that predict the immunotherapy response in PDAC.
Collapse
Affiliation(s)
- Bo Gao
- Department of Hernia and Abdominal Wall, Peking University People's Hospital, Beijing, China
| | - Guohua Hu
- Department of Hernia and Abdominal Wall, Peking University People's Hospital, Beijing, China
| | - Boshi Sun
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenqiang Li
- Department of General Surgery, Jinshan Hospital of Fudan University, Shanghai, China
| | - Hao Yang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
20
|
Sharma A, Becker F, Tao X, Baddela VS, Koczan D, Ludwig C, Vanselow J. Hyperplastic ovarian stromal cells express genes associated to tumor progression: a case study. BMC Vet Res 2024; 20:439. [PMID: 39342193 PMCID: PMC11438404 DOI: 10.1186/s12917-024-04275-6] [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: 11/18/2023] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
The current study presents the analysis of stromal cells obtained from an hyperplastic left-ovary of a Holstein cow. Cultured hyperplastic stromal cells displayed a fibroblast-like morphology and ceased proliferation after the 8th passage. The non-cancerous nature of stromal cells was confirmed by in vitro cell proliferation and migration assays. Negligible amounts of E2 were detected in the spent media of cultured stromal cells, which suggests that stromal cells were non-estradiol synthesizing cells. As revealed in immunofluorescence and gene expression analysis, the hyperplastic stromal cells explicitly expressed vimentin in their cytoskeleton. Upon hematoxylin staining, a highly dense population of stromal cells was observed in the stromal tissue of the hyperplastic ovary. To explore genome-wide alterations, mRNA microarray analysis was performed using Affymetrix Bovine Gene 1.0ST Arrays compared to normal ovarian derived stromal cells. The microarray identified 1396 differentially expressed genes, of which 733 were up- and 663 down-regulated in hyperplastic stromal cells. Importantly, asporin (ASPN) and vascular cell adhesion molecule 1 (VCAM1) were among the highly up-regulated genes. Higher expression of ASPN was also confirmed by immunohistochemistry and RT-qPCR analysis. Ingenuity pathway analysis (IPA) identified about 98 significantly enriched (-log (p value ≥ 1.3) canonical pathways, importantly of which the "Sirutin Signaling Pathway" and "Mitochondrial Dysfunction" were highly activated while "Oxidative phosphorylation" was inhibited. Additionally, higher proportion of hyperplastic stromal cells in the S-phase of cell cycle, could be attributed to higher expression levels of cell proliferation genes such as CCND2 and CDK6.
Collapse
Affiliation(s)
- Arpna Sharma
- Forschungsinstitut für Nutztierbiologie (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, USA.
| | - Frank Becker
- Forschungsinstitut für Nutztierbiologie (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Xuelian Tao
- Forschungsinstitut für Nutztierbiologie (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Vijay Simha Baddela
- Forschungsinstitut für Nutztierbiologie (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Dirk Koczan
- Institut für Immunologie, Universität Rostock, 18055, Rostock, Germany
| | - Carolin Ludwig
- Forschungsinstitut für Nutztierbiologie (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Jens Vanselow
- Forschungsinstitut für Nutztierbiologie (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| |
Collapse
|
21
|
Ohguro H, Watanabe M, Sato T, Nishikiori N, Umetsu A, Higashide M, Yano T, Suzuki H, Miyazaki A, Takada K, Uhara H, Furuhashi M, Hikage F. Application of Single Cell Type-Derived Spheroids Generated by Using a Hanging Drop Culture Technique in Various In Vitro Disease Models: A Narrow Review. Cells 2024; 13:1549. [PMID: 39329734 PMCID: PMC11430518 DOI: 10.3390/cells13181549] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Cell culture methods are indispensable strategies for studies in biological sciences and for drug discovery and testing. Most cell cultures have been developed using two-dimensional (2D) culture methods, but three-dimensional (3D) culture techniques enable the establishment of in vitro models that replicate various pathogenic conditions and they provide valuable insights into the pathophysiology of various diseases as well as more precise results in tests for drug efficacy. However, one difficulty in the use of 3D cultures is selection of the appropriate 3D cell culture technique for the study purpose among the various techniques ranging from the simplest single cell type-derived spheroid culture to the more sophisticated organoid cultures. In the simplest single cell type-derived spheroid cultures, there are also various scaffold-assisted methods such as hydrogel-assisted cultures, biofilm-assisted cultures, particle-assisted cultures, and magnet particle-assisted cultures, as well as non-assisted methods, such as static suspension cultures, floating cultures, and hanging drop cultures. Since each method can be differently influenced by various factors such as gravity force, buoyant force, centrifugal force, and magnetic force, in addition to non-physiological scaffolds, each method has its own advantages and disadvantages, and the methods have different suitable applications. We have been focusing on the use of a hanging drop culture method for modeling various non-cancerous and cancerous diseases because this technique is affected only by gravity force and buoyant force and is thus the simplest method among the various single cell type-derived spheroid culture methods. We have found that the biological natures of spheroids generated even by the simplest method of hanging drop cultures are completely different from those of 2D cultured cells. In this review, we focus on the biological aspects of single cell type-derived spheroid culture and its applications in in vitro models for various diseases.
Collapse
Affiliation(s)
- Hiroshi Ohguro
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (N.N.); (A.U.); (M.H.)
| | - Megumi Watanabe
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (N.N.); (A.U.); (M.H.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.Y.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Nami Nishikiori
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (N.N.); (A.U.); (M.H.)
| | - Araya Umetsu
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (N.N.); (A.U.); (M.H.)
| | - Megumi Higashide
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (N.N.); (A.U.); (M.H.)
| | - Toshiyuki Yano
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.Y.); (M.F.)
| | - Hiromu Suzuki
- Departments of Molecular Biology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan;
| | - Akihiro Miyazaki
- Departments of Oral Surgery, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan;
| | - Kohichi Takada
- Departments of Medical Oncology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan;
| | - Hisashi Uhara
- Departments of Dermatology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan;
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.Y.); (M.F.)
| | - Fumihito Hikage
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (N.N.); (A.U.); (M.H.)
| |
Collapse
|
22
|
Kewitz‐Hempel S, Windisch N, Hause G, Müller L, Sunderkötter C, Gerloff D. Extracellular vesicles derived from melanoma cells induce carcinoma-associated fibroblasts via miR-92b-3p mediated downregulation of PTEN. J Extracell Vesicles 2024; 13:e12509. [PMID: 39315679 PMCID: PMC11420832 DOI: 10.1002/jev2.12509] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
In melanoma, carcinoma-associated fibroblasts (CAFs) are important cellular components in the tumour microenvironment due to their potential to promote tumour growth and metastatic spread of malignant cells. Melanoma cells have the ability to affect non-tumour cells in the microenvironment by releasing extracellular vesicles (EVs). The mechanisms responsible for reprogramming normal dermal fibroblasts (NHDFs) into CAFs remain incompletely understood. However, it is likely thought to be mediated by melanoma-specific miRNAs, which are transported by EVs derived from melanoma cells. Therefore, we wondered if one of the most enriched miRNAs in EVs secreted by melanoma cells, miR-92b-3p, is involved in the conversion of normal fibroblasts into CAFs. We observed that melanoma cell-derived EVs indeed delivered miR-92b-3p into NHDFs and that its accumulation correlated with CAF formation, as demonstrated by enhanced expression of CAF marker genes and increased proliferation and migration. Overexpression of miR-92b-3p in NHDFs revealed similar results, while EVs deficient of miR-92b-3p did not induce a CAF phenotype. As a target we identified PTEN, whose repression led to increased expression of CAF markers. We thus provide a novel pathway of intercellular communication by which melanoma cells control the transformation of CAFs by virtue of EV-transported miRNAs.
Collapse
Affiliation(s)
- Stefanie Kewitz‐Hempel
- Department of Dermatology and VenereologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Nicola Windisch
- Department of Dermatology and VenereologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Gerd Hause
- BiocenterMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Lutz Müller
- Department of Internal Medicine IVHematology and Oncology, Martin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Cord Sunderkötter
- Department of Dermatology and VenereologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Dennis Gerloff
- Department of Dermatology and VenereologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| |
Collapse
|
23
|
Xiong Y, Xu X, Zhou X, Tong Y, Yu C. Anlotinib inhibits cervical cancer cell proliferation and invasion by suppressing cytokine secretion in activated cancer-associated fibroblasts. Front Oncol 2024; 14:1412660. [PMID: 39193386 PMCID: PMC11347301 DOI: 10.3389/fonc.2024.1412660] [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: 04/05/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Objective The aim of this study was to investigate whether anlotinib could exert an inhibitory effect on the proliferation and invasion of cervical cancer cells by inhibiting cytokines secreted by activated cancer-associated fibroblasts (CAFs). Methods CAFs were isolated from cervical cancer tissues and experimentally studied in vivo and in vitro. Molecular biology experimental methods were used to verify whether anlotinib could inhibit the pro-carcinogenic effects of CAFs derived from cervical cancer tissues. Results CAFs promote the proliferation and invasion of cervical cancer cells. Anlotinib inhibited the activation of CAFs and suppressed the promotion of cervical cancer cells by CAFs. Anlotinib inhibited the expression of multiple cytokines within CAFs and suppressed the release of interleukin (IL)-6 (IL-6) and IL-8. In vivo studies have shown that anlotinib diminished the growth of xenografted cervical cancer cells, and treatment in combination with docetaxel had an even more significant tumor growth inhibitory effect. Conclusion Anlotinib inhibits the pro-cancer effects of CAFs by suppressing the activation of CAFs and the secretion of pro-cancer cytokines. Our findings suggest that the combination of anlotinib and docetaxel may be a potential strategy for the treatment of refractory cervical cancer.
Collapse
Affiliation(s)
- Yaozu Xiong
- Department of Radiation Oncology, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Xiaoting Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xilei Zhou
- Department of Radiation Oncology, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Yusuo Tong
- Department of Radiation Oncology, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Changhua Yu
- Department of Radiation Oncology, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| |
Collapse
|
24
|
Chen CY, Yang SH, Chang PY, Chen SF, Nieh S, Huang WY, Lin YC, Lee OKS. Cancer-Associated-Fibroblast-Mediated Paracrine and Autocrine SDF-1/CXCR4 Signaling Promotes Stemness and Aggressiveness of Colorectal Cancers. Cells 2024; 13:1334. [PMID: 39195225 PMCID: PMC11352219 DOI: 10.3390/cells13161334] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/20/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer mortality worldwide, and cancer-associated fibroblasts (CAFs) play a major role in the tumor microenvironment (TME), which facilitates the progression of CRC. It is critical to understand how CAFs promote the progression of CRC for the development of novel therapeutic approaches. The purpose of this study was to understand how CAF-derived stromal-derived factor-1 (SDF-1) and its interactions with the corresponding C-X-C motif chemokine receptor 4 (CXCR4) promote CRC progression. Our study focused on their roles in promoting tumor cell migration and invasion and their effects on the characteristics of cancer stem cells (CSCs), which ultimately impact patient outcomes. Here, using in vivo approaches and clinical histological samples, we analyzed the influence of secreted SDF-1 on CRC progression, especially in terms of tumor cell behavior and stemness. We demonstrated that CAF-secreted SDF-1 significantly enhanced CRC cell migration and invasion through paracrine signaling. In addition, the overexpression of SDF-1 in CRC cell lines HT29 and HCT-116 triggered these cells to generate autocrine SDF-1 signaling, which further enhanced their CSC characteristics, including those of migration, invasion, and spheroid formation. An immunohistochemical study showed a close relationship between SDF-1 and CXCR4 expression in CRC tissue, and this significantly affected patient outcomes. The administration of AMD3100, an inhibitor of CXCR4, reversed the entire phenomenon. Our results strongly suggest that targeting this signaling axis in CRC is a feasible approach to attenuating tumor progression, and it may, therefore, serve as an alternative treatment method to improve the prognosis of patients with CRC, especially those with advanced, recurrent, or metastatic CRC following standard therapy.
Collapse
Affiliation(s)
- Chao-Yang Chen
- Division of Colon and Rectal Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Shih-Hsien Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan;
- Office of General Affairs and Occupational Safety, National Defense Medical Center, Taipei 11490, Taiwan
| | - Ping-Ying Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Su-Feng Chen
- Department of Dentistry, School of Dentistry, China Medical University, Taichung 40433, Taiwan;
| | - Shin Nieh
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Wen-Yen Huang
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yu-Chun Lin
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Oscar Kuang-Sheng Lee
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei 11211, Taiwan
- Department of Orthopedics, China Medical University Hospital, Taichung 40402, Taiwan
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung 40402, Taiwan
| |
Collapse
|
25
|
Tan J, Zhu L, Shi J, Zhang J, Kuang J, Guo Q, Zhu X, Chen Y, Zhou C, Gao X. Evaluation of drug resistance for EGFR-TKIs in lung cancer via multicellular lung-on-a-chip. Eur J Pharm Sci 2024; 199:106805. [PMID: 38763450 DOI: 10.1016/j.ejps.2024.106805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/10/2024] [Accepted: 05/17/2024] [Indexed: 05/21/2024]
Abstract
Drug resistance to irreversible epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) is a primary factor affecting their therapeutic efficacy in human non-small cell lung cancer (NSCLC). NSCLC cells can undergo epithelial-mesenchymal transition (EMT) induced by many factors in the tumour microenvironment (TME), which plays a crucial role in tumour drug resistance. In this study, a multicellular lung-on-a-chip that can realise the cell co-culture of the human non-small cell lung cancer cell line HCC827, human foetal lung fibroblasts (HFL-1), and human umbilical vein endothelial cells (HUVECs) is prepared. The TME was simulated on the chip combined with perfusion and other factors, and the drug evaluation of osimertinib was performed to explore the drug resistance mechanism of EGFR-TKIs. In the early stages, a two-dimensional static cell co-culture was achieved by microchip, and the results showed that HFL-1 cells could be transformed into cancer-associated fibroblasts (CAFs), and HCC827 cells could undergo EMT, both of which were mediated by Interleukin-6 (IL-6). Vimentin (VIM) and Alpha Skeletal Muscle Actin (a-SMA) expression of HFL-1 was upregulated, whereas E-cadherin (E-cad) expression of HCC827 was down-regulated. Further, N-cadherin (N-cad) expression of HCC827 was upregulated. In both the static cell co-culture and multicellular lung-on-a-chip, HCC827 cells with CAFs co-culture or IL-6 treatment developed resistance to osimertinib. Further use of the IL-6 antibody inhibitor tocilizumab could reverse EGFR-TKI resistance to a certain extent. Combination therapy with tocilizumab and EGFR-TKIs may provide a novel therapeutic strategy for overcoming EGFR-TKI resistance caused by EMT in NSCLC. Furthermore, the lung-on-a-chip can simulate complex TME and can be used for evaluating tumour resistance and exploring mechanisms, with the potential to become an important tool for personalised diagnosis, treatment, and biomedical research.
Collapse
Affiliation(s)
- Jianfeng Tan
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510030, China
| | - Leqing Zhu
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China; Shenzhen Clinical Medical College, Southern Medical University, Shenzhen,518101, China
| | - Jingyan Shi
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Jianhua Zhang
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Jun Kuang
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Quanwei Guo
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Xiaojia Zhu
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Yuliang Chen
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Chengbin Zhou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510030, China; Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, China.
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
26
|
Inoue C, Miki Y, Saito-Koyama R, Okada Y, Sasano H, Suzuki T. Dipeptidyl peptidase 4-positive cancer-associated fibroblasts enhance lung adenocarcinoma growth. Pathol Res Pract 2024; 260:155418. [PMID: 38908333 DOI: 10.1016/j.prp.2024.155418] [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: 02/01/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of fibroblasts with various features in the cancer stroma and have been reported to influence cancer progression through cell-cell interactions in various types of malignancies, including lung adenocarcinoma (LUAD). Dipeptidyl peptidase 4 (DPP4) is a transmembrane protein with serine protease activity and is involved in the progression of tumors, metabolic diseases, and autoimmune diseases. In the present study, we focused on the role of DPP4-positive CAFs in LUAD. Immunohistochemistry revealed that 38 of 89 LUAD patients showed DPP4 expression in the fibrous stroma, and patients harboring DPP4-positive CAFs were more often male, had a higher Brinkman index, and had a higher Ki-67 labeling index of tumor cells than those with DPP4-negative CAFs. DPP4-positivity was associated with the expression of other CAF markers, α-SMA, periostin, and podoplanin, as well as a cellular senescence marker, p16. In the in vitro study, conditioned media collected from pulmonary fibroblast (OUS-11, HPF, and HPF-C)-induced overexpression of DPP4 significantly promoted the proliferation of LUAD cells (A549 and PC-9) and increased the expression levels of MCP-1, IL-8, IL-6, and GCSF in the media compared to those in controls. In addition, OUS-11 overexpression in DPP4 overexpression increased periostin expression. In conclusion, DPP4-positive CAFs could promote lung adenocarcinoma cell growth by producing soluble factors, and DPP4 inhibition may inhibit cancer progression.
Collapse
Affiliation(s)
- Chihiro Inoue
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan.
| | - Yasuhiro Miki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan
| | - Ryoko Saito-Koyama
- Department of Pathology, National Hospital Organization, Sendai Medical Center, Sendai, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan
| | - Takashi Suzuki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan
| |
Collapse
|
27
|
Lin J, Lu W, Huang B, Yang W, Wang X. The role of tissue-derived extracellular vesicles in tumor microenvironment. Tissue Cell 2024; 89:102470. [PMID: 39002287 DOI: 10.1016/j.tice.2024.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
The tumor microenvironment (TME) is a highly heterogeneous ecosystem that plays critical roles in the initiation, progression, invasion, and metastasis of cancers. Extracellular vesicles (EVs), as emerging components of the host-tumor communication, are lipid-bilayer membrane structures that are secreted by most cell types into TEM and increasingly recognized as critical elements that regulate the interaction between tumor cells and their surroundings. They contain a variety of bioactive molecules, such as proteins, nucleic acids, and lipids, and participate in various pathophysiological processes while regulating intercellular communication. While many studies have focused on the EVs derived from different body fluids or cell culture supernatants, the direct isolation of tissue-derived EVs (Ti-EVs) has garnered more attention due to the advantages of tissue specificity and accurate reflection of tissue microenvironment. In this review, we summarize the protocol for isolating Ti-EVs from different tissue interstitium, discuss the role of tumor-derived and adipose tissue-derived Ti-EVs in regulating TME. In addition, we sum up the latest application of Ti-EVs as potential biomarkers for cancer diseases.
Collapse
Affiliation(s)
- Jin Lin
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wan Lu
- Jiangxi Provincial Key Laboratory of Birth Defect for Prevention and Control, Medical Genetics Center, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Weiming Yang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
28
|
Whitman MA, Mantri M, Spanos E, Estroff LA, De Vlaminck I, Fischbach C. Bone mineral density affects tumor growth by shaping microenvironmental heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604333. [PMID: 39091735 PMCID: PMC11291034 DOI: 10.1101/2024.07.19.604333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Breast cancer bone metastasis is the leading cause of mortality in patients with advanced breast cancer. Although decreased mineral density is a known risk factor for bone metastasis, the underlying mechanisms remain poorly understood because studying the isolated effect of bone mineral density on tumor heterogeneity is challenging with conventional approaches. Here, we investigate how bone mineral content affects tumor growth and microenvironmental complexity in vivo by combining single-cell RNA-sequencing with mineral-containing or mineral-free decellularized bone matrices. We discover that the absence of bone mineral significantly influences fibroblast and immune cell heterogeneity, promoting phenotypes that increase tumor growth and alter the response to injury or disease. Importantly, we observe that the stromal response to matrix mineral content depends on host immunocompetence and the murine tumor model used. Collectively, our findings suggest that bone mineral density affects tumor growth by altering microenvironmental complexity in an organism-dependent manner.
Collapse
Affiliation(s)
- Matthew A. Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14850
| | - Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14850
| | - Emmanuel Spanos
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14850
| | - Lara A. Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14850
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14850
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14850
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14850
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14850
| |
Collapse
|
29
|
Adapa SR, Sami A, Meshram P, Ferreira GC, Jiang RHY. Uncovering Porphyrin Accumulation in the Tumor Microenvironment. Genes (Basel) 2024; 15:961. [PMID: 39062740 PMCID: PMC11275590 DOI: 10.3390/genes15070961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Heme, an iron-containing tetrapyrrole, is essential in almost all organisms. Heme biosynthesis needs to be precisely regulated particularly given the potential cytotoxicity of protoporphyrin IX, the intermediate preceding heme formation. Here, we report on the porphyrin intermediate accumulation within the tumor microenvironment (TME), which we propose to result from dysregulation of heme biosynthesis concomitant with an enhanced cancer survival dependence on mid-step genes, a process we recently termed "Porphyrin Overdrive". Specifically, porphyrins build up in both lung cancer cells and stromal cells in the TME. Within the TME's stromal cells, evidence supports cancer-associated fibroblasts (CAFs) actively producing porphyrins through an imbalanced pathway. Conversely, normal tissues exhibit no porphyrin accumulation, and CAFs deprived of tumor cease porphyrin overproduction, indicating that both cancer and tumor-stromal porphyrin overproduction is confined to the cancer-specific tissue niche. The clinical relevance of our findings is implied by establishing a correlation between imbalanced porphyrin production and overall poorer survival in more aggressive cancers. These findings illuminate the anomalous porphyrin dynamics specifically within the tumor microenvironment, suggesting a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Swamy R. Adapa
- USF Genomics Program, Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
| | - Abdus Sami
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.S.); (G.C.F.)
| | - Pravin Meshram
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
| | - Gloria C. Ferreira
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.S.); (G.C.F.)
- Department of Chemistry, College of Arts and Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Rays H. Y. Jiang
- USF Genomics Program, Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA;
| |
Collapse
|
30
|
Meng X, Liu Z, Deng L, Yang Y, Zhu Y, Sun X, Hao Y, He Y, Fu J. Hydrogen Therapy Reverses Cancer-Associated Fibroblasts Phenotypes and Remodels Stromal Microenvironment to Stimulate Systematic Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401269. [PMID: 38757665 PMCID: PMC11267370 DOI: 10.1002/advs.202401269] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/02/2024] [Indexed: 05/18/2024]
Abstract
Tumor microenvironment (TME) plays an important role in the tumor progression. Among TME components, cancer-associated fibroblasts (CAFs) show multiple tumor-promoting effects and can induce tumor immune evasion and drug-resistance. Regulating CAFs can be a potential strategy to augment systemic anti-tumor immunity. Here, the study observes that hydrogen treatment can alleviate intracellular reactive oxygen species of CAFs and reshape CAFs' tumor-promoting and immune-suppressive phenotypes. Accordingly, a controllable and TME-responsive hydrogen therapy based on a CaCO3 nanoparticles-coated magnesium system (Mg-CaCO3) is developed. The hydrogen therapy by Mg-CaCO3 can not only directly kill tumor cells, but also inhibit pro-tumor and immune suppressive factors in CAFs, and thus augment immune activities of CD4+ T cells. As implanted in situ, Mg-CaCO3 can significantly suppress tumor growth, turn the "cold" primary tumor into "hot", and stimulate systematic anti-tumor immunity, which is confirmed by the bilateral tumor transplantation models of "cold tumor" (4T1 cells) and "hot tumor" (MC38 cells). This hydrogen therapy system reverses immune suppressive phenotypes of CAFs, thus providing a systematic anti-tumor immune stimulating strategy by remodeling tumor stromal microenvironment.
Collapse
Affiliation(s)
- Xiaoyan Meng
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Liang Deng
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| | - Yangzi Yang
- Department of Orthopedic SurgerySpine CenterChangzheng HospitalNavy Medical UniversityNo. 415 Fengyang RoadShanghai200003P. R. China
| | - Yingchun Zhu
- Key Laboratory of Inorganic Coating MaterialsShanghai Institute of CeramicsChinese Academy of SciencesShanghai200050P. R. China
| | - Xiaoying Sun
- College of SciencesShanghai UniversityShanghai200444P. R. China
| | - Yongqiang Hao
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Jingke Fu
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| |
Collapse
|
31
|
Zhu J, Zhang K, Chen Y, Ge X, Wu J, Xu P, Yao J. Progress of single-cell RNA sequencing combined with spatial transcriptomics in tumour microenvironment and treatment of pancreatic cancer. J Transl Med 2024; 22:563. [PMID: 38867230 PMCID: PMC11167806 DOI: 10.1186/s12967-024-05307-3] [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: 11/27/2023] [Accepted: 05/16/2024] [Indexed: 06/14/2024] Open
Abstract
In recent years, single-cell analyses have revealed the heterogeneity of the tumour microenvironment (TME) at the genomic, transcriptomic, and proteomic levels, further improving our understanding of the mechanisms of tumour development. Single-cell RNA sequencing (scRNA-seq) technology allow analysis of the transcriptome at the single-cell level and have unprecedented potential for exploration of the characteristics involved in tumour development and progression. These techniques allow analysis of transcript sequences at higher resolution, thereby increasing our understanding of the diversity of cells found in the tumour microenvironment and how these cells interact in complex tumour tissue. Although scRNA-seq has emerged as an important tool for studying the tumour microenvironment in recent years, it cannot be used to analyse spatial information for cells. In this regard, spatial transcriptomics (ST) approaches allow researchers to understand the functions of individual cells in complex multicellular organisms by understanding their physical location in tissue sections. In particular, in related research on tumour heterogeneity, ST is an excellent complementary approach to scRNA-seq, constituting a new method for further exploration of tumour heterogeneity, and this approach can also provide unprecedented insight into the development of treatments for pancreatic cancer (PC). In this review, based on the methods of scRNA-seq and ST analyses, research progress on the tumour microenvironment and treatment of pancreatic cancer is further explained.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated Yangzhou University, Jiangsu Province, China
| | - Ke Zhang
- Dalian Medical University, Dalian, China
| | - Yuan Chen
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated Yangzhou University, Jiangsu Province, China
| | - Xinyu Ge
- Dalian Medical University, Dalian, China
| | - Junqing Wu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated Yangzhou University, Jiangsu Province, China
| | - Peng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated Yangzhou University, Jiangsu Province, China.
| | - Jie Yao
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated Yangzhou University, Jiangsu Province, China.
| |
Collapse
|
32
|
De Palma M, Hanahan D. Milestones in tumor vascularization and its therapeutic targeting. NATURE CANCER 2024; 5:827-843. [PMID: 38918437 DOI: 10.1038/s43018-024-00780-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 04/22/2024] [Indexed: 06/27/2024]
Abstract
Research into the mechanisms and manifestations of solid tumor vascularization was launched more than 50 years ago with the proposition and experimental demonstrations that angiogenesis is instrumental for tumor growth and was, therefore, a promising therapeutic target. The biological knowledge and therapeutic insights forthcoming have been remarkable, punctuated by new concepts, many of which were not foreseen in the early decades. This article presents a perspective on tumor vascularization and its therapeutic targeting but does not portray a historical timeline. Rather, we highlight eight conceptual milestones, integrating initial discoveries and recent progress and posing open questions for the future.
Collapse
Affiliation(s)
- Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| |
Collapse
|
33
|
Rademaekers M, Johansson EO, Johansson E, Roberg K, Wiechec E. Tumor-matched and unmatched cancer associated fibroblasts exhibit differential effect on proliferation and FMOD and MMP9 gene expression in head and neck squamous cell carcinoma cells when cocultured in spheroids. Cancer Cell Int 2024; 24:190. [PMID: 38822309 PMCID: PMC11143562 DOI: 10.1186/s12935-024-03388-0] [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: 12/19/2023] [Accepted: 05/25/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are the major cellular component of the tumor microenvironment and are known to affect tumor growth and response to various treatments. This study was undertaken to investigate the crosstalk between tumor-matched or unmatched CAFs and head and neck squamous cell carcinoma (HNSCC) cells regarding tumor growth and treatment response. METHODS Three HNSCC cell lines (LK0412, LK0902 and LK0923), were cocultured in 2D or in 3D with their tumor-matched CAFs, site matched CAFs from other tumors or normal oral fibroblasts (NOFs). Cell proliferation was assessed as the amount of Ki67 positive cells/ spheroid area in formalin-fixed- paraffin-embedded 3D spheroids stained with Ki67 antibody. Viability after seven days of cisplatin treatment was measured with CellTiter-Glo 3D Viability Assay. The mRNA expression of CAF-associated markers (ACTA2, COL1A2, FAP, PDGFRα, PDGFRβ, PDPN, POSTN and S100A4) in CAFs before and after coculture with tumor cells as well as mRNA expression of CAF-induced genes (MMP1, MMP9 and FMOD) in tumor cells separated from CAFs after co-culture was measured with RT-qPCR. The expression of selected protein biomarkers was validated with immunohistochemistry based on previous mRNA expression results. RESULTS The proliferation of the LK0412 and LK0902 tumor spheroids varied significantly when cocultured with different CAFs and NOFs as shown by Ki-67 positive cells. RT‒qPCR analysis revealed different molecular profile of the analyzed HNSCC-derived CAFs concerning the expression of CAF-associated markers. The interaction between CAFs and HNSCC cells was more pronounced after coculture with unmatched CAFs as shown by changes in mRNA expression pattern of CAF-specific markers. Additionally, the unmatched CAFs significantly upregulated the mRNA expression of MMP1, MMP9 and FMOD in tumor cells compared to tumor-matched CAFs. CONCLUSION Our results indicate that tumor-matched CAFs are unique for each tumor and affect the proliferation and the gene/protein expression of tumor cells in a distinct manner. The interaction between tumor unmatched CAFs and HNSCC cells in the tumor spheroids is associated with significant changes in the mRNA expression of CAF-specific markers and significant increases in FMOD and MMP9 in tumor cells compared to when cocultured with tumor-matched CAFs. Taken together, our results show how important the selection of CAFs is to get a reliable in vitro model that mimics the patients' tumor.
Collapse
Affiliation(s)
- Max Rademaekers
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Otorhinolaryngology, Region Östergötland, Linköping, Sweden
| | - Emil Oliver Johansson
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ellen Johansson
- Department of Otorhinolaryngology, Region Östergötland, Linköping, Sweden
| | - Karin Roberg
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
- Department of Otorhinolaryngology, Region Östergötland, Linköping, Sweden.
| | - Emilia Wiechec
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
34
|
Milosevic V, Östman A. Interactions between cancer-associated fibroblasts and T-cells: functional crosstalk with targeting and biomarker potential. Ups J Med Sci 2024; 129:10710. [PMID: 38863724 PMCID: PMC11165253 DOI: 10.48101/ujms.v129.10710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 06/13/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population recognized as a key component of the tumour microenvironment (TME). Cancer-associated fibroblasts are known to play an important role in maintaining and remodelling the extracellular matrix (ECM) in the tumour stroma, supporting cancer progression and inhibiting the immune system's response against cancer cells. This review aims to summarize the immunomodulatory roles of CAFs, particularly focussing on their T-cell suppressive effects. Cancer-associated fibroblasts have several ways by which they can affect the tumour's immune microenvironment (TIME). For example, their interactions with macrophages and dendritic cells (DCs) create an immunosuppressive milieu that can indirectly affect T-cell anticancer immunity and enable immune evasion. In addition, a number of recent studies have confirmed CAF-mediated direct suppressive effects on T-cell anticancer capacity through ECM remodelling, promoting the expression of immune checkpoints, cytokine secretion and the release of extracellular vesicles. The consequential impact of CAFs on T-cell function is then reflected in affecting T-cell proliferation and apoptosis, migration and infiltration, differentiation and exhaustion. Emerging evidence highlights the existence of specific CAF subsets with distinct capabilities to modulate the immune landscape of TME in various cancers, suggesting the possibility of their exploitation as possible prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Vladan Milosevic
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Arne Östman
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
Masucci M, Karlsson C, Blomqvist L, Ernberg I. Bridging the Divide: A Review on the Implementation of Personalized Cancer Medicine. J Pers Med 2024; 14:561. [PMID: 38929782 PMCID: PMC11204735 DOI: 10.3390/jpm14060561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/05/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
The shift towards personalized cancer medicine (PCM) represents a significant transformation in cancer care, emphasizing tailored treatments based on the genetic understanding of cancer at the cellular level. This review draws on recent literature to explore key factors influencing PCM implementation, highlighting the role of innovative leadership, interdisciplinary collaboration, and coordinated funding and regulatory strategies. Success in PCM relies on overcoming challenges such as integrating diverse medical disciplines, securing sustainable investment for shared infrastructures, and navigating complex regulatory landscapes. Effective leadership is crucial for fostering a culture of innovation and teamwork, essential for translating complex biological insights into personalized treatment strategies. The transition to PCM necessitates not only organizational adaptation but also the development of new professional roles and training programs, underscoring the need for a multidisciplinary approach and the importance of team science in overcoming the limitations of traditional medical paradigms. The conclusion underscores that PCM's success hinges on creating collaborative environments that support innovation, adaptability, and shared vision among all stakeholders involved in cancer care.
Collapse
Affiliation(s)
- Michele Masucci
- Department of Learning, Informatics, Management and Ethics (LIME), Karolinska Institutet, Tomtebodavägen 18B, 171 65 Solna, Sweden
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Claes Karlsson
- Department of Oncology-Pathology (Onc-Pat), Karolinska Institutet, Anna Steckséns gata 30A, D2:04, 171 65 Solna, Sweden;
| | - Lennart Blomqvist
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Anna Steckséns gata 53, 171 65 Solna, Sweden;
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| |
Collapse
|
36
|
Sánchez-Ramírez D, Mendoza-Rodríguez MG, Alemán OR, Candanedo-González FA, Rodríguez-Sosa M, Montesinos-Montesinos JJ, Salcedo M, Brito-Toledo I, Vaca-Paniagua F, Terrazas LI. Impact of STAT-signaling pathway on cancer-associated fibroblasts in colorectal cancer and its role in immunosuppression. World J Gastrointest Oncol 2024; 16:1705-1724. [PMID: 38764833 PMCID: PMC11099434 DOI: 10.4251/wjgo.v16.i5.1705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/28/2024] [Accepted: 04/01/2024] [Indexed: 05/09/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the most commonly diagnosed and deadliest types of cancer worldwide. CRC displays a desmoplastic reaction (DR) that has been inversely associated with poor prognosis; less DR is associated with a better prognosis. This reaction generates excessive connective tissue, in which cancer-associated fibroblasts (CAFs) are critical cells that form a part of the tumor microenvironment. CAFs are directly involved in tumorigenesis through different mechanisms. However, their role in immunosuppression in CRC is not well understood, and the precise role of signal transducers and activators of transcription (STATs) in mediating CAF activity in CRC remains unclear. Among the myriad chemical and biological factors that affect CAFs, different cytokines mediate their function by activating STAT signaling pathways. Thus, the harmful effects of CAFs in favoring tumor growth and invasion may be modulated using STAT inhibitors. Here, we analyze the impact of different STATs on CAF activity and their immunoregulatory role.
Collapse
Affiliation(s)
- Damián Sánchez-Ramírez
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Mónica G Mendoza-Rodríguez
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Omar R Alemán
- Department of Biology, Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Ciudad Universitaria, Mexico City 04510, Mexico
| | - Fernando A Candanedo-González
- Department of Pathology, National Medical Center Century XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Miriam Rodríguez-Sosa
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Juan José Montesinos-Montesinos
- Laboratorio de Células Troncales Mesenquimales, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Mauricio Salcedo
- Unidad de Investigacion en Biomedicina y Oncologia Genomica, Instituto Mexciano del Seguro Social, Mexico City 07300, Mexico
| | - Ismael Brito-Toledo
- Servicio de Colon y Recto, Hospital de Oncología Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Felipe Vaca-Paniagua
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Luis I Terrazas
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| |
Collapse
|
37
|
You S, Han X, Xu Y, Sui L, Song K, Yao Q. High expression of SLC7A1 in high-grade serous ovarian cancer promotes tumor progression and is involved in MAPK/ERK pathway and EMT. Cancer Med 2024; 13:e7217. [PMID: 38752472 PMCID: PMC11097251 DOI: 10.1002/cam4.7217] [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: 11/29/2023] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024] Open
Abstract
Our previous studies have shown that upregulation of SLC7A1 in epithelial ovarian cancer (EOC) tumor cells significantly increases cancer cell proliferation, migration, and cisplatin resistance; however, the molecular mechanism by which SLC7A1 functions in EOC remains unknown. In later studies, we found that SLC7A1 is also highly expressed in the interstitial portion of high-grade serous ovarian cancer (HGSOC), but the significance of this high expression in the interstitial remains unclear. Here, we showed the Interstitial high expression of SLC7A1 in HGSOC by immunohistochemistry. SLC7A1 enriched in cancer-associated fibroblasts (CAFs) was upregulated by TGF-β1. Transwell assay, scratch assay, cck8 assay and cell adhesion assay showed that SLC7A1 highly expressed in CAFs promoted tumor cells invasion, migration and metastasis in vitro. The effect of SLC7A1 on MAPK and EMT pathway proteins in ovarian cancer (OC) was verified by RNA sequencing and western blotting. Overexpression of SLC7A1 in OC is involved in MAPK/ ERK pathway and EMT. In general, in HGSOC, CAFs overexpressing SLC7A1 supported the migration and invasion of tumor cells; SLC7A1 is highly expressed in ovarian cancer and is involved in ERK phosphorylation and EMT signaling in MAPK signaling pathway. This suggests that SLC7A1 may be a potential therapeutic target for OC metastasis.
Collapse
Affiliation(s)
- Shijing You
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Xiahui Han
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Yuance Xu
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Lei Sui
- Department of Gynecological OncologyAffiliated Qingdao Central Hospital of Qingdao UniversityQingdaoShandongChina
| | - Kejuan Song
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Qin Yao
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
38
|
Qin R, Ma X, Pu S, Shen C, Hu D, Liu C, Wang K, Wang Y. Identification and validation of a signature based on myofibroblastic cancer-associated fibroblast marker genes for predicting prognosis, immune infiltration, and therapeutic response in bladder cancer. Investig Clin Urol 2024; 65:263-278. [PMID: 38714517 PMCID: PMC11076800 DOI: 10.4111/icu.20230300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/08/2023] [Accepted: 01/02/2024] [Indexed: 05/10/2024] Open
Abstract
PURPOSE Myofibroblastic cancer-associated fibroblasts (myCAFs) are important components of the tumor microenvironment closely associated with tumor stromal remodeling and immunosuppression. This study aimed to explore myCAFs marker gene biomarkers for clinical diagnosis and therapy for patients with bladder cancer (BC). MATERIALS AND METHODS BC single-cell RNA sequencing (scRNA-seq) data were obtained from the National Center for Biotechnology Information Sequence Read Archive. Transcriptome and clinical data were downloaded from The Cancer Genome Atlas and the Gene Expression Omnibus databases. Subsequently, univariate Cox and LASSO (Least Absolute Shrinkage and Selection Operator regression) regression analyses were performed to construct a prognostic signature. Immune cell activity was estimated using single-sample gene set enrichment analysis whilst the TIDE (tumor immune dysfunction and exclusion) method was employed to assess patient response to immunotherapy. The chemotherapy response of patients with BC was evaluated using genomics of drug sensitivity in cancer. Furthermore, Immunohistochemistry was used to verify the correlation between MAP1B expression and immunotherapy efficacy. The scRNA-seq data were analyzed to identify myCAFs marker genes. RESULTS Combined with bulk RNA-sequencing data, we constructed a two-gene (COL6A1 and MAP1B) risk signature. In patients with BC, the signature demonstrated outstanding prognostic value, immune infiltration, and immunotherapy response. This signature served as a crucial guide for the selection of anti-tumor chemotherapy medications. Additionally, immunohistochemistry confirmed that MAP1B expression was significantly correlated with immunotherapy efficacy. CONCLUSIONS Our findings revealed a typical prognostic signature based on myCAF marker genes, which offers patients with BC a novel treatment target alongside theoretical justification.
Collapse
Affiliation(s)
- Ruize Qin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaocheng Ma
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shi Pu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengquan Shen
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ding Hu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changxue Liu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kongjia Wang
- Department of Urology, Qingdao Municipal Hospital, Qingdao, China
| | - Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
39
|
Yimin E, Lu C, Zhu K, Li W, Sun J, Ji P, Meng M, Liu Z, Yu C. Function and mechanism of exosomes derived from different cells as communication mediators in colorectal cancer metastasis. iScience 2024; 27:109350. [PMID: 38500820 PMCID: PMC10945197 DOI: 10.1016/j.isci.2024.109350] [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] [Indexed: 03/20/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the second leading cause of cancer-related mortality, with metastasis being the primary determinant of poor prognosis in patients. Investigating the molecular mechanisms underlying CRC metastasis is currently a prominent and challenging area of research. Exosomes, as crucial intercellular communication mediators, facilitate the transfer of metabolic and genetic information from cells of origin to recipient cells. Their roles in mediating information exchange between CRC cells and immune cells, fibroblasts, and other cell types are pivotal in reshaping the tumor microenvironment, regulating key biological processes such as invasion, migration, and formation of pre-metastatic niche. This article comprehensively examines the communication function and mechanism of exosomes derived from different cells in cancer metastasis, while also presenting an outlook on current research advancements and future application prospects. The aim is to offer a distinctive perspective that contributes to accurate diagnosis and rational treatment strategies for CRC.
Collapse
Affiliation(s)
- Yimin E
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing 210011, Jiangsu, China
| | - Chen Lu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Long Mian Avenue 109 Jiangning, Nanjing 211112, Jiangsu, China
| | - Kuixuan Zhu
- Department of Radiotherapy, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming 650100, Yunan, China
| | - Wenyuan Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing 210011, Jiangsu, China
| | - Jing Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing 210011, Jiangsu, China
| | - Pengcheng Ji
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Long Mian Avenue 109 Jiangning, Nanjing 211112, Jiangsu, China
| | - Minjie Meng
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing 210011, Jiangsu, China
| | - Zhengxia Liu
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing 210011, Jiangsu, China
| | - Chunzhao Yu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Long Mian Avenue 109 Jiangning, Nanjing 211112, Jiangsu, China
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing 210011, Jiangsu, China
| |
Collapse
|
40
|
Pietras K, Sjölund J. Cellular plasticity in the breast cancer ecosystem. Ups J Med Sci 2024; 129:10629. [PMID: 38571887 PMCID: PMC10989214 DOI: 10.48101/ujms.v129.10629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024] Open
Abstract
The complex interplay between genetically diverse tumor cells and their microenvironment significantly influences cancer progression and therapeutic responses. This review highlights recent findings on cellular plasticity and heterogeneity within the breast cancer ecosystem, focusing on the roles of cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). We discuss evidence suggesting that breast cancer cells exhibit phenotypic plasticity driven by both intrinsic genetic factors and external microenvironmental cues, impacting treatment responses and disease recurrence. Moreover, single-cell RNA sequencing studies reveal diverse subtypes of CAFs and TAMs, each with distinct functional gene expression programs and spatial organization within the tumor microenvironment. Understanding the hierarchical relationships and niche cues governing cellular phenotypes offers new opportunities for targeted therapeutic interventions. By elucidating the organizational principles of the tumor ecosystem, future therapies may target phenotypic states or entire cellular niches, advancing precision medicine approaches in breast cancer treatment.
Collapse
Affiliation(s)
- Kristian Pietras
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Jonas Sjölund
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| |
Collapse
|
41
|
Gu Q, Ma Z, Wang Q, Dai Y, Shi W, Jiao Z. Knockout of Shcbp1 sensitizes immunotherapy by regulating α-SMA positive cancer-associated fibroblasts. Mol Carcinog 2024; 63:601-616. [PMID: 38169303 DOI: 10.1002/mc.23675] [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: 08/14/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/05/2024]
Abstract
The crucial role of cancer-associated fibroblasts (CAFs) in promoting T-cell exclusion has a significant impact on tumor immune evasion and resistance to immunotherapy. Therefore, enhancing T-cell infiltration into solid tumors has emerged as a pivotal area of research. We achieved a conventional knockout of Shcbp1 (Shcbp1-/- ) through CRISPR/Cas9 gene editing and crossed these mice with spontaneous breast cancer MMTV-PyMT mice, resulting in PyMT Shcbp1-/- mice. The different CAF subtypes were detected by flow cytometry analysis (FCA). We evaluated collagen and CAFs levels using Sirius red staining, immunohistochemistry (IHC), and immunofluorescence (IF). Primary tumor cells and CAFs were isolated from both PyMT Shcbp1+/+ and PyMT Shcbp1-/- mice. We analyzed CAFs' proliferation, invasion, migration, apoptosis, and cell cycle. Transwell coculture experiments were performed with primary tumor cells and CAFs to evaluate the role of CAFs in increasing the sensitivity of tumor cells to Erdafitinib. Tumors from PyMT Shcbp1+/+ and PyMT Shcbp1-/- mice were orthotopically transplanted to assess the therapeutic effect of the Erdafitinib and PD-1 combination. CAFs and T-cell infiltration in these tumors were assessed using FCA and IF. Knockout of Shcbp1 leads to a significant reduction in tumor burden, promotes longer survival, and decreases CAFs in MMTV-PyMT. Moreover, knockout of Shcbp1 enhances the sensitivity of Erdafitinib, leading to effective inhibition of CAFs' proliferation and invasion, as well as the induction of apoptosis. Additionally, it results in cell cycle arrest at the G2/M phase in vitro. Meanwhile, Shcbp1-/- CAFs change the sensitivity of Shcbp1-/- tumor cells to Erdafitinib compared to Shcbp1+/+ CAFs. Importantly, knockout of Shcbp1 boosts the effectiveness of Erdafitinib in combination with immune checkpoint blockade therapy by augmenting T-cell infiltration through CAFs regulation in vivo. Our findings demonstrate that knockout of Shcbp1 holds significant potential in enhancing the therapeutic response of Erdafitinib combined with PD-1 antibody treatment, offering promising prospects for future breast cancer therapies.
Collapse
Affiliation(s)
- Qianlin Gu
- The Second Clinical Medical College, Lanzhou University, Lanzhou city, Gansu Province, China
| | - Zhijian Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou city, Gansu Province, China
| | - Qiaoyan Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou city, Gansu Province, China
| | - Yiwei Dai
- The Second Clinical Medical College, Lanzhou University, Lanzhou city, Gansu Province, China
| | - Wengui Shi
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou city, Gansu Province, China
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou city, Gansu Province, China
| | - Zuoyi Jiao
- The Second Clinical Medical College, Lanzhou University, Lanzhou city, Gansu Province, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou city, Gansu Province, China
- Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou city, Gansu Province, China
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou city, Gansu Province, China
| |
Collapse
|
42
|
Godina C, Belting M, Vallon-Christersson J, Isaksson K, Bosch A, Jernström H. Caveolin-1 gene expression provides additional prognostic information combined with PAM50 risk of recurrence (ROR) score in breast cancer. Sci Rep 2024; 14:6675. [PMID: 38509243 PMCID: PMC10954762 DOI: 10.1038/s41598-024-57365-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/18/2024] [Indexed: 03/22/2024] Open
Abstract
Combining information from the tumor microenvironment (TME) with PAM50 Risk of Recurrence (ROR) score could improve breast cancer prognostication. Caveolin-1 (CAV1) is a marker of an active TME. CAV1 is a membrane protein involved in cell signaling, extracellular matrix organization, and tumor-stroma interactions. We sought to investigate CAV1 gene expression in relation to PAM50 subtypes, ROR score, and their joint prognostic impact. CAV1 expression was compared between PAM50 subtypes and ROR categories in two cohorts (SCAN-B, n = 5326 and METABRIC, n = 1980). CAV1 expression was assessed in relation to clinical outcomes using Cox regression and adjusted for clinicopathological predictors. Effect modifications between CAV1 expression and ROR categories on clinical outcome were investigated using multiplicative and additive two-way interaction analyses. Differential gene expression and gene set enrichment analyses were applied to compare high and low expressing CAV1 tumors. All samples expressed CAV1 with the highest expression in the Normal-like subtype. Gene modules consistent with epithelial-mesenchymal transition (EMT), hypoxia, and stromal activation were associated with high CAV1 expression. CAV1 expression was inversely associated with ROR category. Interactions between CAV1 expression and ROR categories were observed in both cohorts. High expressing CAV1 tumors conferred worse prognosis only within the group classified as ROR high. ROR gave markedly different prognostic information depending on the underlying CAV1 expression. CAV1, a potential mediator between the malignant cells and TME, could be a useful biomarker that enhances and further refines PAM50 ROR risk stratification in patients with ROR high tumors and a potential therapeutic target.
Collapse
Affiliation(s)
- Christopher Godina
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Barngatan 4, 221 85, Lund, Sweden.
| | - Mattias Belting
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Barngatan 4, 221 85, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Skåne, Sweden
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Vallon-Christersson
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Barngatan 4, 221 85, Lund, Sweden
| | - Karolin Isaksson
- Department of Clinical Sciences Lund, Surgery, Lund University and Kristianstad Hospital, Kristianstad, Sweden
| | - Ana Bosch
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Barngatan 4, 221 85, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Skåne, Sweden
| | - Helena Jernström
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Barngatan 4, 221 85, Lund, Sweden.
| |
Collapse
|
43
|
Leng X, Liu J, Jin A, Zheng H, Wu J, Zhong L, Li Q, Li D. Multi-omics Analyses Reveal Function of Apolipoprotein E in Alternative Splicing and Tumor Immune Microenvironment in Kidney Renal Clear Cell Carcinoma via Pan-cancer Analysis. Cell Biochem Biophys 2024; 82:1-13. [PMID: 38182861 DOI: 10.1007/s12013-023-01211-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 12/22/2023] [Indexed: 01/07/2024]
Abstract
Apolipoprotein E (APOE) regulates lipid metabolism, associated with the development of various cancers. However, its precise prognostic significance and functions in alternative splicing and the tumor immune microenvironment remain unclear. In this study, we extracted APOE expression in pan-cancer from TCGA and analyzed mRNA transcriptome, cell lines, and protein levels. Furthermore, we analyzed the alternative splicing expression of the APOE gene transcript with prognostic profiles using the OncoSplicing database. We obtained 73 common APOE genes to perform functional enrichment analysis, assess the correlation between genes and immune cells using TIMER, EPIC, and ssGSEA methods, and examine the prognostic significance using the UALCAN database. Finally, single-cell data was employed to assess the correlation between APOE genes and cell functions. Our findings revealed that APOE expression varies across different tumor types and cancer cell lines. The alternative splicing analysis demonstrated that APOE transcript expression levels have prognostic value in cancers such as LGG, KIRC, and KIRP. Functional enrichment analysis indicated significant associations between APOE and various immune cells, such as macrophages, CD8 T cells, and NK cells, with significant implications for prognosis. Moreover, single-cell data indicated that APOE was primarily expressed in renal epithelial cells among stromal cells and in macrophages among immune cells, significantly negatively correlated with five functional states. Our study represents the first comprehensive exploration of APOE's function in pan-cancers and identifies APOE as a potential biomarker in cancer pathogenesis, prognosis, and immune therapeutic target.
Collapse
Affiliation(s)
- Xin Leng
- Department of Urology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, China
| | - Jianhu Liu
- Department of Urology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, China
| | - Anqi Jin
- The BioBank, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, China
| | - Hongfang Zheng
- Department of Urology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, China
| | - Jiulong Wu
- Department of Urology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, China
| | - Longfei Zhong
- Department of Urology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, China
| | - Qiaoxin Li
- Department of Urology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, 215300, China
| | - Dongfeng Li
- Department of Urology, The Third People's Hospital of Kunshan, Suzhou, 215300, China.
| |
Collapse
|
44
|
Wei R, Song J, Liu X, Huo S, Liu C, Liu X. Immunosuppressive MFAP2 + cancer associated fibroblasts conferred unfavorable prognosis and therapeutic resistance in gastric cancer. Cell Oncol (Dordr) 2024; 47:55-68. [PMID: 37540308 DOI: 10.1007/s13402-023-00849-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 08/05/2023] Open
Abstract
PURPOSE To explore the predictive merit of MFAP2+ cancer associated fibroblasts (CAFs) infiltration for clinical outcomes and adjuvant chemotherapy or immunotherapy responsiveness in gastric cancer (GC). METHODS In this study, several independent cohorts were included respectively to dissect the relationship of clinical outcomes, therapeutic responses and tumor microenvironment with different MFAP2+ CAFs infiltration. Drug sensitivity analysis was conducted to predict the relationship between MFAP2+ CAFs infiltration and targeted drug response. Kaplan-Meier curves and the log-rank test were used to compare clinical outcomes of patients with different MFAP2+ CAFs infiltration. RESULTS High MFAP2+ CAFs infiltration yielded inferior prognosis in terms of overall survival, progress free survival and recurrence free survival in GC. Patients with low MFAP2+ CAFs infiltration were more likely to gain benefit from adjuvant therapy. Moreover, low MFAP2+ CAFs infiltration could predict a promising response to immunotherapy in GC patients. MFAP2+ CAFs with immunosuppressive features were highly relevant to immune evasive contexture characterized by the dysfunction of CD8+ T cells. We found that MFAP2+ CAFs communicated with T cells, B cells and Macrophages through releasing macrophage migration inhibitor factor (MIF), which further suggested that MFAP2+ CAFs might promote therapeutic resistance through regulating T cells dysfunction and M2 macrophages polarization. CONCLUSION Immunosuppressive MFAP2+ CAFs constructed an immune evasive tumor microenvironment characterized by incapacitated immune effector cells, consequently predicting inferior clinical outcomes and response on adjuvant therapy and immunotherapy in patients with GC. The potential of immunosuppressive MFAP2+ CAFs as a therapeutic target for GC deserved thoroughly exploration.
Collapse
Affiliation(s)
- Rongyuan Wei
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Junquan Song
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xuanjun Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shiying Huo
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Xiaowen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
45
|
Chen Z, Miao Y, Tan Z, Hu Q, Wu Y, Li X, Guo W, Gu J. scCancer2: data-driven in-depth annotations of the tumor microenvironment at single-level resolution. Bioinformatics 2024; 40:btae028. [PMID: 38243719 PMCID: PMC10868330 DOI: 10.1093/bioinformatics/btae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024] Open
Abstract
SUMMARY Single-cell RNA-seq (scRNA-seq) is a powerful technique for decoding the complex cellular compositions in the tumor microenvironment (TME). As previous studies have defined many meaningful cell subtypes in several tumor types, there is a great need to computationally transfer these labels to new datasets. Also, different studies used different approaches or criteria to define the cell subtypes for the same major cell lineages. The relationships between the cell subtypes defined in different studies should be carefully evaluated. In this updated package scCancer2, designed for integrative tumor scRNA-seq data analysis, we developed a supervised machine learning framework to annotate TME cells with annotated cell subtypes from 15 scRNA-seq datasets with 594 samples in total. Based on the trained classifiers, we quantitatively constructed the similarity maps between the cell subtypes defined in different references by testing on all the 15 datasets. Secondly, to improve the identification of malignant cells, we designed a classifier by integrating large-scale pan-cancer TCGA bulk gene expression datasets and scRNA-seq datasets (10 cancer types, 175 samples, 663 857 cells). This classifier shows robust performances when no internal confidential reference cells are available. Thirdly, scCancer2 integrated a module to process the spatial transcriptomic data and analyze the spatial features of TME. AVAILABILITY AND IMPLEMENTATION The package and user documentation are available at http://lifeome.net/software/sccancer2/ and https://doi.org/10.5281/zenodo.10477296.
Collapse
Affiliation(s)
- Zeyu Chen
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yuxin Miao
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing 100084, China
| | - Zhiyuan Tan
- Department of Finance, Shanghai Advanced Institute of Finance, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qifan Hu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yanhong Wu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xinqi Li
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing 100084, China
| | - Wenbo Guo
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing 100084, China
| |
Collapse
|
46
|
Padathpeedika Khalid J, Mary Martin T, Prathap L, Abhimanyu Nisargandha M, Boopathy N, Kishore Kumar MS. Exploring Tumor-Promoting Qualities of Cancer-Associated Fibroblasts and Innovative Drug Discovery Strategies With Emphasis on Thymoquinone. Cureus 2024; 16:e53949. [PMID: 38468988 PMCID: PMC10925941 DOI: 10.7759/cureus.53949] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
Tumor epithelial development and chemoresistance are highly promoted by the tumor microenvironment (TME), which is mostly made up of the cancer stroma. This is due to several causes. Cancer-associated fibroblasts (CAFs) stand out among them as being essential for the promotion of tumors. Understanding the fibroblastic population within a single tumor is made more challenging by the undeniable heterogeneity within it, even though particular stromal alterations are still up for debate. Numerous chemical signals released by tumors improve the connections between heterotypic fibroblasts and CAFs, promoting the spread of cancer. It becomes essential to have a thorough understanding of this complex microenvironment to effectively prevent solid tumor growth. Important new insights into the role of CAFs in the TME have been revealed by recent studies. The objective of this review is to carefully investigate the relationship between CAFs in tumors and plant secondary metabolites, with a focus on thymoquinone (TQ). The literature published between 2010 and 2023 was searched in PubMed and Google Scholar with keywords such as TQ, TME, cancer-associated fibroblasts, mechanism of action, and flavonoids. The results showed a wealth of data substantiating the activity of plant secondary metabolites, particularly TQ's involvement in blocking CAF operations. Scrutinized research also clarified the wider effect of flavonoids on pathways related to cancer. The present study highlights the complex dynamics of the TME and emphasizes the critical role of CAFs. It also examines the possible interventions provided by secondary metabolites found in plants, with TQ playing a vital role in regulating CAF function based on recent literature.
Collapse
Affiliation(s)
- Jabir Padathpeedika Khalid
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Taniya Mary Martin
- Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Lavanya Prathap
- Department of Anatomy, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Milind Abhimanyu Nisargandha
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Nisha Boopathy
- Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Meenakshi Sundaram Kishore Kumar
- Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
47
|
Lyu SI, Johannsen J, Simon AG, Knipper K, Wuerdemann N, Sharma SJ, Thelen M, Hansen KK, Fretter C, Klasen C, Esser J, Suchan MC, Abing H, Zimmermann PH, Schultheis AM, Schloesser HA, Klussmann JP, Quaas A, Eckel HNC. Co-expression patterns of cancer associated fibroblast markers reveal distinct subgroups related to patient survival in oropharyngeal squamous cell carcinoma. Front Cell Dev Biol 2024; 12:1337361. [PMID: 38328551 PMCID: PMC10847231 DOI: 10.3389/fcell.2024.1337361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024] Open
Abstract
Background: The incidence of oropharyngeal squamous cell carcinoma (OPSCC) is rapidly increasing in high income countries due to its association with persistent high-risk human papilloma virus (HPV) infection. Recent scientific advances have highlighted the importance of the tumor microenvironment in OPSCC. In this study, including 216 OPSCC patients, we analyze the composition of four established markers of cancer associated fibroblasts (CAFs) in the context of intratumoral CD8 T-cell infiltration. Methods: Immunohistochemical staining for fibroblast activation protein (FAP), platelet-derived growth factor receptor beta (PDGFRb), periostin, alpha smooth muscle actin (α-SMA) and CD8 were analyzed digitally and their association with survival, tumor- and patient characteristics was assessed. Results: Co-expression of CAF markers was frequent but not associated with HPV status. FAPhigh and PDGFRbhigh expression were associated with increased CD8 T-cell infiltration. Low expression of PDGFRb improved patient survival in female patients but not in male patients. We identified PDGFRblow periostinlow α-SMAlow status as an independent predictor of improved survival (hazard ratio 0.377, p = 0.006). Conclusion: These findings elucidate the co-expression of four established CAF markers in OPSCC and underscore their association with T-cell infiltration and patient survival. Future analyses of CAF subgroups in OPSCC may enable the development of individualized therapies.
Collapse
Affiliation(s)
- Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Jannik Johannsen
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Karl Knipper
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Nora Wuerdemann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Shachi Jenny Sharma
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Martin Thelen
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Kevin Karl Hansen
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Caroline Fretter
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Charlotte Klasen
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Julia Esser
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Malte Christian Suchan
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Helen Abing
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Philipp Heinrich Zimmermann
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Anne Maria Schultheis
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Hans Anton Schloesser
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Jens Peter Klussmann
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Hans Nikolaus Caspar Eckel
- Faculty of Medicine and University Hospital of Cologne, Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
48
|
Han H, Qian C, Song M, Zhong C, Zhao Y, Lu Y. Fibroblasts: invigorated targets in pre-metastatic niche formation. Int J Biol Sci 2024; 20:1110-1124. [PMID: 38322116 PMCID: PMC10845297 DOI: 10.7150/ijbs.87680] [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: 07/01/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
At present, tumor metastasis still remains the leading contributor to high recurrence and mortality in cancer patients. There have been no clinically effective therapeutic strategies for treating patients with metastatic cancer. In recent years, a growing body of evidence has shown that the pre-metastatic niche (PMN) plays a crucial role in driving tumor metastasis. Nevertheless, a clear and detailed understanding of the formation of PMN is still lacking given the fact that PMN formation involves in a wealth of complicated communications and underlying mechanisms between primary tumors and metastatic target organs. Despite that the roles of numerous components including tumor exosomes and extracellular vesicles in influencing the evolution of PMN have been well documented, the involvement of cancer-associated fibroblasts (CAFs) in the tumor microenvironment for controlling PMN formation is frequently overlooked. It has been increasingly recognized that fibroblasts trigger the formation of PMN by virtue of modulating exosomes, metabolism and so on. In this review, we mainly summarize the underlying mechanisms of fibroblasts from diverse origins in exerting impacts on PMN evolution, and further highlight the prospective strategies for targeting fibroblasts to prevent PMN formation.
Collapse
Affiliation(s)
- Hongkuan Han
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengyao Song
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chongjin Zhong
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Zhao
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
49
|
Fergatova A, Affara NI. The cellular triumvirate: fibroblasts entangled in the crosstalk between cancer cells and immune cells. Front Immunol 2024; 14:1337333. [PMID: 38313431 PMCID: PMC10835808 DOI: 10.3389/fimmu.2023.1337333] [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: 11/12/2023] [Accepted: 12/29/2023] [Indexed: 02/06/2024] Open
Abstract
This review article will focus on subpopulations of fibroblasts that get reprogrammed by tumor cells into cancer-associated fibroblasts. Throughout this article, we will discuss the intricate interactions between fibroblasts, immune cells, and tumor cells. Unravelling complex intercellular crosstalk will pave the way for new insights into cellular mechanisms underlying the reprogramming of the local tumor immune microenvironment and propose novel immunotherapy strategies that might have potential in harnessing and modulating immune system responses.
Collapse
|
50
|
Bagger MM, Sjölund J, Kim J, Kohler KT, Villadsen R, Jafari A, Kassem M, Pietras K, Rønnov-Jessen L, Petersen OW. Evidence of steady-state fibroblast subtypes in the normal human breast as cells-of-origin for perturbed-state fibroblasts in breast cancer. Breast Cancer Res 2024; 26:11. [PMID: 38229104 PMCID: PMC10790388 DOI: 10.1186/s13058-024-01763-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Human breast cancer most frequently originates within a well-defined anatomical structure referred to as the terminal duct lobular unit (TDLU). This structure is endowed with its very own lobular fibroblasts representing one out of two steady-state fibroblast subtypes-the other being interlobular fibroblasts. While cancer-associated fibroblasts (CAFs) are increasingly appreciated as covering a spectrum of perturbed states, we lack a coherent understanding of their relationship-if any-with the steady-state fibroblast subtypes. To address this, we here established two autologous CAF lines representing inflammatory CAFs (iCAFs) and myofibroblast CAFs (myCAFs) and compared them with already established interlobular- and lobular fibroblasts with respect to their origin and impact on tumor formation. METHODS Primary breast tumor-derived CAFs were transduced to express human telomerase reverse transcriptase (hTERT) and sorted into CD105low and CD105high populations using fluorescence-activated cell sorting (FACS). The two populations were tested for differentiation similarities to iCAF and myCAF states through transcriptome-wide RNA-Sequencing (RNA-Seq) including comparison to an available iCAF-myCAF cell state atlas. Inference of origin in interlobular and lobular fibroblasts relied on RNA-Seq profiles, immunocytochemistry and growth characteristics. Osteogenic differentiation and bone formation assays in culture and in vivo were employed to gauge for origin in bone marrow-derived mesenchymal stem cells (bMSCs). Functional characteristics were assessed with respect to contractility in culture and interaction with tumor cells in mouse xenografts. The cells' gene expression signatures were tested for association with clinical outcome of breast cancer patients using survival data from The Cancer Genome Atlas database. RESULTS We demonstrate that iCAFs have properties in common with interlobular fibroblasts while myCAFs and lobular fibroblasts are related. None of the CAFs qualify as bMSCs as revealed by lack of critical performance in bone formation assays. Functionally, myCAFs and lobular fibroblasts are almost equally tumor promoting as opposed to iCAFs and interlobular fibroblasts. A myCAF gene signature is found to associate with poor breast cancer-specific survival. CONCLUSIONS We propose that iCAFs and myCAFs originate in interlobular and lobular fibroblasts, respectively, and more importantly, that the tumor-promoting properties of lobular fibroblasts render the TDLU an epicenter for breast cancer evolution.
Collapse
Affiliation(s)
- Mikkel Morsing Bagger
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden.
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Jiyoung Kim
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - René Villadsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Laboratory of Molecular Endocrinology, KMEB, Department of Endocrinology, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Lone Rønnov-Jessen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ole William Petersen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|