1
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
2
|
Saeed Issa B, Adhab AH, Salih Mahdi M, Kyada A, Ganesan S, Bhanot D, Naidu KS, Kaur S, Mansoor AS, Radi UK, Saadoun Abd N, Kariem M. Decoding the complex web: cellular and molecular interactions in the lung tumour microenvironment. J Drug Target 2025; 33:666-690. [PMID: 39707828 DOI: 10.1080/1061186x.2024.2445772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
The lung tumour microenvironment (TME) or stroma is a dynamic space of numerous cells and their released molecules. This complicated web regulates tumour progression and resistance to different modalities. Lung cancer cells in conjunction with their stroma liberate a wide range of factors that dampen antitumor attacks by innate immunity cells like natural killer (NK) cells and also adaptive responses by effector T cells. These factors include numerous growth factors, exosomes and epigenetic regulators, and also anti-inflammatory cytokines. Understanding the intricate interactions between tumour cells and various elements within the lung TME, such as immune and stromal cells can help provide novel strategies for better management and treatment of lung malignancies. The current article discusses the complex network of cells and signalling molecules, which mediate communications in lung TME. By elucidating these multifaceted interactions, we aim to provide insights into potential therapeutic targets and strategies for lung cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sharnjeet Kaur
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | | | - Usama Kadem Radi
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Nasr Saadoun Abd
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
3
|
Zhang T, Hu Y, Li H, Wang J, Xu Q, Xu Y, Sun H. Stage pT0-T1 rectal cancers: emphasis on submucosal high intensity on high-resolution T2-weighted imaging and other morphological features. Acta Radiol 2025; 66:558-566. [PMID: 39988912 DOI: 10.1177/02841851251316435] [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: 02/25/2025]
Abstract
BackgroundIdentification and staging of rectal cancer are mainly based on the difference in signal intensity (SI) between the tumor and normal structures of the intestinal wall on T2-weighted imaging. However, differentiating stage pT0-T1 from pT2 rectal tumors is difficult using routine magnetic resonance imaging (MRI) sequences.PurposeTo summarize and explore whether MRI findings from routine imaging can help differentiate pT0-T1 from pT2 rectal tumors.Material and MethodsA total of 110 patients with pT0-T2 rectal cancer underwent preoperative pelvic MRI examinations and tumor resection without preoperative chemoradiotherapy. MRI findings of rectal lesions (including tumor location, shape, longitudinal length, maximum cross-section, submucosal high intensity [SHI], extramural fibrotic scarring, wall shrinkage, lesion-to-wall signal intensity ratio, and presence of lymph node with short axis over 3 mm) and clinical characteristics were analyzed by univariate and multivariate analyses to screen the independent factors associated with pathological results.ResultsOf all the lesions, 32 tumors were proved to be pT0-T1 and 78 tumors were pT2. Univariate and multivariate logistic regression analyses revealed that tumor shape (odds ratio [OR] = 24.607, P < 0.001), SHI (OR = 6.129, P = 0.002), and extramural fibrotic scarring (OR = 0.110, P = 0.007) were independent factors distinguishing pT0-T1 tumors from pT2 tumors. If the rectal lesion has a cauliflower-like shape with SHI and no extramural fibrotic scarring, it is more likely to be a pT0-T1 tumor.ConclusionThe imaging features obtained from the routine MRI sequence showed potential value for differentiating pT0-T1 from pT2 rectal tumors.
Collapse
Affiliation(s)
- Tongyin Zhang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, PR China
- Graduate School, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, PR China
| | - Yuwan Hu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, PR China
- Graduate School, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, PR China
| | - Haoyu Li
- Department of Radiology, China-Japan Friendship Hospital, Beijing, PR China
- Graduate School, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, PR China
| | - Juan Wang
- Department of Radiology, Civil Aviation General Hospital, Beijing, PR China
| | - Qiaoyu Xu
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China
| | - Yanyan Xu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, PR China
| | - Hongliang Sun
- Department of Radiology, China-Japan Friendship Hospital, Beijing, PR China
| |
Collapse
|
4
|
Li C, Liao J, Chen B, Wang Q. Heterogeneity of the tumor immune cell microenvironment revealed by single-cell sequencing in head and neck cancer. Crit Rev Oncol Hematol 2025; 209:104677. [PMID: 40023465 DOI: 10.1016/j.critrevonc.2025.104677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
Head and neck cancer (HNC) is the sixth most common disease in the world. The recurrence rate of patients is relatively high, and the heterogeneity of tumor immune microenvironment (TIME) cells may be an important reason for this. Single-cell sequencing (SCS) is currently the most promising and mature application in cancer research. It can identify unique genes expressed in cells and study tumor heterogeneity. According to current research, the heterogeneity of immune cells has become an important factor affecting the occurrence and development of HNC. SCSs can provide effective therapeutic targets and prognostic factors for HNC patients through analyses of gene expression levels and cell heterogeneity. Therefore, this study analyzes the basic theory of HNC and the development of SCS technology, elaborating on the application of SCS technology in HNC and its potential value in identifying HNC therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Jia Liao
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Bo Chen
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan 629000, China.
| |
Collapse
|
5
|
Li Y, Zhou M, Hu X, Xie T, Peng W, Zhang L, Tang M, Hu R, He Y. Cancer-associated fibroblast-derived exosomal FAM83F regulates KIF23 expression to promote the malignant progression and reduce radiosensitivity in non-small cell lung cancer. Cytotechnology 2025; 77:50. [PMID: 39867833 PMCID: PMC11759729 DOI: 10.1007/s10616-025-00713-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) have been shown to play a crucial role in the progression of non-small cell lung cancer (NSCLC). Exosomes derived from CAFs have emerged as important mediators of intercellular communication in the tumor microenvironment, contributing to cancer progression. Therefore, it is essential to further investigate the mechanisms by which CAF-derived exosomes regulate NSCLC. CAFs promoted NSCLC cell proliferation, invasion, and migration, while also suppressing radiosensitivity. We observed an upregulation of FAM83F expression in both NSCLC cells and NSCLC cells treated with conditioned medium from CAFs. Notably, CAF-derived exosomes were found to transfer FAM83F to NSCLC cells, thereby enhancing the malignant properties of the cancer cells. In contrast, FAM83F-deficient CAF-derived exosomes exerted inhibitory effects on NSCLC cell proliferation, invasion, and migration, while also sensitizing the cells to radiotherapy. FAM83F was found to interact with KIF23 in NSCLC cells, and the overexpression of KIF23 attenuated the effects induced by FAM83F-deficient exosomes in NSCLC cells. Moreover, FAM83F-deficient CAF-derived exosomes were effective in inhibiting tumor formation in vivo. Our findings highlight the crucial role of CAF-derived exosomal FAM83F in promoting NSCLC progression and conferring resistance to radiotherapy. Targeting this signaling pathway may offer promising therapeutic strategies for combating NSCLC progression and improving patient outcomes. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00713-x.
Collapse
Affiliation(s)
- Yi Li
- Department of Thoracic Surgery, Suining Central Hospital, Suining, 629099 China
| | - Mingming Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing University Cancer Hospital, Chongqing, 400030 China
| | - Xiaogang Hu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing University Cancer Hospital, Chongqing, 400030 China
| | - Tingting Xie
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing University Cancer Hospital, Chongqing, 400030 China
| | - Wenli Peng
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing University Cancer Hospital, Chongqing, 400030 China
| | - Lina Zhang
- Department Radiotherapy, Affiliated Hospital of Hebei University, Hebei, 071000 China
| | - Minxin Tang
- Department of Medical Laboratory, Chongqing Institute of Orthopedics of Traditional Chinese Medicine, Chongqing Orthopedic Hospital of Traditional Chinese Medicine, The People’s Hospital of Chongqing Yuzhong District, Chongqing, 400039 China
| | - Rui Hu
- Department of Medical Laboratory, Chongqing Institute of Orthopedics of Traditional Chinese Medicine, Chongqing Orthopedic Hospital of Traditional Chinese Medicine, The People’s Hospital of Chongqing Yuzhong District, Chongqing, 400039 China
| | - Yongpeng He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute, Chongqing Cancer Hospital, Chongqing University Cancer Hospital, Chongqing, 400030 China
| |
Collapse
|
6
|
Palmer EP, Cronise KE, Haines LA, Das S, Offermann A, Easton CP, Regan DP. Osteosarcoma Exosome Priming of Primary Human Lung Fibroblasts Induces an Immune Modulatory and Protumorigenic Phenotype. CANCER RESEARCH COMMUNICATIONS 2025; 5:594-608. [PMID: 40099972 PMCID: PMC11987067 DOI: 10.1158/2767-9764.crc-24-0371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/07/2024] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
SIGNIFICANCE These findings provide a critical first step in characterizing the capacity of OS-derived exosomes to reprogram primary LFs toward a tumor-promoting inflammatory phenotype in vitro, offering novel molecular targets for the modulation of fibroblasts in the lung microenvironment as potential therapeutic strategies to prevent OS metastasis.
Collapse
Affiliation(s)
- Eric P. Palmer
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Kathryn E. Cronise
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Laurel A. Haines
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Sunetra Das
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Aaron Offermann
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Carina P. Easton
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Daniel P. Regan
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
7
|
Liu S, Jin P. Advances and Challenges in 3D Bioprinted Cancer Models: Opportunities for Personalized Medicine and Tissue Engineering. Polymers (Basel) 2025; 17:948. [PMID: 40219336 PMCID: PMC11991528 DOI: 10.3390/polym17070948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Cancer is the second leading cause of death worldwide, after cardiovascular disease, claiming not only a staggering number of lives but also causing considerable health and economic devastation, particularly in less-developed countries. Therapeutic interventions are impeded by differences in patient-to-patient responses to anti-cancer drugs. A personalized medicine approach is crucial for treating specific patient groups and includes using molecular and genetic screens to find appropriate stratifications of patients who will respond (and those who will not) to treatment regimens. However, information on which risk stratification method can be used to hone in on cancer types and patients who will be likely responders to a specific anti-cancer agent remains elusive for most cancers. Novel developments in 3D bioprinting technology have been widely applied to recreate relevant bioengineered tumor organotypic structures capable of mimicking the human tissue and microenvironment or adequate drug responses in high-throughput screening settings. Parts are autogenously printed in the form of 3D bioengineered tissues using a computer-aided design concept where multiple layers include different cell types and compatible biomaterials to build specific configurations. Patient-derived cancer and stromal cells, together with genetic material, extracellular matrix proteins, and growth factors, are used to create bioprinted cancer models that provide a possible platform for the screening of new personalized therapies in advance. Both natural and synthetic biopolymers have been used to encourage the growth of cells and biological materials in personalized tumor models/implants. These models may facilitate physiologically relevant cell-cell and cell-matrix interactions with 3D heterogeneity resembling real tumors.
Collapse
Affiliation(s)
- Sai Liu
- Health Science Center, Yangtze University, Jingzhou 434023, China;
| | | |
Collapse
|
8
|
Shah DD, Chorawala MR, Raghani NR, Patel R, Fareed M, Kashid VA, Prajapati BG. Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med Oncol 2025; 42:117. [PMID: 40102282 DOI: 10.1007/s12032-025-02641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Tumor microenvironment (TME) denotes the non-cancerous cells and components presented in the tumor, including molecules produced and released by them. Interactions between cancer cells, immune cells, stromal cells, and the extracellular matrix within the TME create a dynamic ecosystem that can either promote or hinder tumor growth and spread. The TME plays a pivotal role in either promoting or inhibiting tumor growth and dissemination, making it a critical factor to consider in the development of effective cancer therapies. Understanding the intricate interplay within the TME is crucial for devising effective cancer therapies. Combination therapies involving inhibitors of immune checkpoint blockade (ICB), and/or chemotherapy now offer new approaches for cancer therapy. However, it remains uncertain how to best utilize these strategies in the context of the complex tumor microenvironment. Oncogene-driven changes in tumor cell metabolism can impact the TME to limit immune responses and present barriers to cancer therapy. Cellular and acellular components in tumor microenvironment can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Components in the TME can reprogram tumor behavior and influence responses to treatments, facilitating immune evasion, nutrient deprivation, and therapeutic resistance. Moreover, the TME can influence angiogenesis, promoting the formation of blood vessels that sustain tumor growth. Notably, the TME facilitates immune evasion, establishes a nutrient-deprived milieu, and induces therapeutic resistance, hindering treatment efficacy. A paradigm shift from a cancer-centric model to a TME-centric one has revolutionized cancer research and treatment. However, effectively targeting specific cells or pathways within the TME remains a challenge, as the complexity of the TME poses hurdles in designing precise and effective therapies. This review highlights challenges in targeting the tumor microenvironment to achieve therapeutic efficacy; explore new approaches and technologies to better decipher the tumor microenvironment; and discuss strategies to intervene in the tumor microenvironment and maximize therapeutic benefits.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Rajanikant Patel
- Department of Product Development, Granules Pharmaceuticals Inc., 3701 Concorde Parkway, Chantilly, VA, 20151, USA
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 13713, Riyadh, Saudi Arabia
| | - Vivekanand A Kashid
- MABD Institute of Pharmaceutical Education and Research, Babhulgaon, Yeola, Nashik, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
9
|
Lo Cicero A, Campora S, Lo Buglio G, Cinà P, Lo Pinto M, Scilabra SD, Ghersi G. Enhancing therapeutic efficacy through degradation of endogenous extracellular matrix in primary breast tumor spheroids. FEBS J 2025. [PMID: 40098313 DOI: 10.1111/febs.70069] [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: 07/18/2024] [Revised: 11/08/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Solid tumors have a complex extracellular matrix (ECM) that significantly affects tumor behavior and response to therapy. Understanding the ECM's role is crucial for advancing cancer research and treatment. This study established an in vitro model using primary cells isolated from a rat breast tumor to generate three-dimensional spheroids. Monolayer cells and spheroid cultures exhibited different protein expression patterns, with primary tumor spheroids presenting an increased level of ECM-related proteins and a more complex extracellular environment. Furthermore, spheroids produce endogenous collagen type I matrix, which is the main component of the tumoral ECM. This matrix is arranged predominantly around the 3D structure, mimicking the conditions of solid tumors. Treatments with recombinant collagenases class II (acting on the linear collagen region) and class I (acting on the 3D-helix region) completely degrade collagen within the spheroid structure. Collagenase pretreatment enhances the accessibility of the anticancer drug doxorubicin to penetrate the core of spheroids and sensitize them to doxorubicin-induced cytotoxicity. Our findings highlight the importance of overcoming drug resistance in breast cancer by targeting the ECM and proposing a novel strategy for improving therapeutic outcomes in solid tumors. By employing a three-dimensional spheroid model, with an endogenous ECM, we can offer more relevant insights into tumor biology and treatment responses.
Collapse
Affiliation(s)
- Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Biomedical Engineering Bioscience Center of the University of Cincinnati, OH, USA
| | - Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Pharmacy, University of Copenhagen, Denmark
| | | | - Margot Lo Pinto
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Simone Dario Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Abiel Srl, Palermo, Italy
| |
Collapse
|
10
|
Zhou D, Li X, Liu W, Zhang M, Cheng Y, Xu Z, Gao J, Wang Y. A novel approach for engineering DHCM/GelMA microgels: application in hepatocellular carcinoma cell encapsulation and chemoresistance research. Front Bioeng Biotechnol 2025; 13:1564543. [PMID: 40161518 PMCID: PMC11949893 DOI: 10.3389/fbioe.2025.1564543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Liver cancer, a highly aggressive malignancy, continues to present significant challenges in therapeutic management due to its pronounced chemoresistance. This resistance, which undermines the efficacy of conventional chemotherapy and targeted therapies, is driven by multifaceted mechanisms, with increasing emphasis placed on the protective role of the tumor microenvironment (TME). The hepatocellular carcinoma extracellular matrix (ECM), a primary non-cellular component of the TME, has emerged as a critical regulator in cancer progression and drug resistance, particularly in hepatocellular carcinoma cell (HCC). In this study, a hybrid biomimetic hydrogel was engineered by integrating decellularized hepatocellular carcinoma matrix (DHCM) with gelatin methacrylate (GelMA) precursors. This composite DHCM/GelMA hydrogel was designed to replicate the physicochemical and functional properties of the hepatocellular carcinoma ECM, thereby offering a biomimetic platform to explore the interactions between HCCs and their microenvironment. Leveraging a custom-designed microfluidic 3D printing platform, we achieved high-throughput fabrication of HCC-encapsulated DHCM/GelMA microgels, characterized by enhanced uniformity, biocompatibility, and scalability. These microgels facilitated the construction of hepatocellular carcinoma microtissues, which were subsequently employed for chemoresistance studies. Our findings revealed that DHCM/GelMA microgels closely mimic the hepatocellular carcinoma tumor microenvironment, effectively recapitulating key features of ECM-mediated drug resistance. Mechanistic studies further demonstrated that DHCM significantly upregulates the expression of Aquaporin 3 (AQP3) in the encapsulated HCCs. This upregulation potentially activates mTOR signaling-associated autophagy pathways, thereby enhancing chemoresistance in HCCs. These biomimetic models provide a robust and versatile platform for studying the underlying mechanisms of drug resistance and evaluating therapeutic interventions. This innovative approach highlights the potential of DHCM/GelMA microgels as a transformative tool in cancer-associated tissue engineering and anticancer drug screening. By enabling detailed investigations into the role of ECM in chemoresistance, this study contributes to advancing therapeutic research and offers promising strategies to overcome drug resistance, ultimately improving clinical outcomes in liver cancer treatment.
Collapse
Affiliation(s)
- Dandan Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Geriatric Medicine, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Xiaoxiao Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wencun Liu
- Department of Radiology, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Mingjun Zhang
- Department of Clinical Laboratory, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Ying Cheng
- Department of Clinical Laboratory, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Zhousong Xu
- Department of Clinical Laboratory, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Jian Gao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyang Wang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2025; 33:295-315. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
12
|
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
|
13
|
Yao L, Zhou C, Liu L, He J, Wang Y, Wang A. Cancer-associated fibroblasts promote growth and dissemination of esophageal squamous cell carcinoma cells by secreting WNT family member 5A. Mol Cell Biochem 2025:10.1007/s11010-025-05223-0. [PMID: 39954174 DOI: 10.1007/s11010-025-05223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common and aggressive subtype of esophageal cancer. This research investigates the functions of cancer-associated fibroblasts (CAFs) in the malignant phenotype of ESCC and probes the underpinning mechanism. Key CAF-associated proteins in ESCC were identified using bioinformatics analyses. ESCC cell lines were co-cultured with CAFs, followed by the addition of neutralizing antibodies against WNT family member 5A (WNT5A) (Anti-WNT5A; AW) and frizzled class receptor 5 (FZD5) (Anti-FZD5; AF), or a human recombinant protein of WNT5A (rWNT5A; rW). The effects of CAF stimulation and the neutralizing or recombinant proteins on the growth and dissemination of ESCC cells were investigated. In addition, ESCC cells were transplanted into nude mice for in vivo assessment of tumor growth and metastasis. WNT5A was identified as a CAF-associated protein linked to poor prognosis in ESCC. Co-culturing with CAFs augmented proliferation, mobility, and apoptosis resistance of ESCC cells. These effects were negated by the AW or AF but restored by rW. WNT5A interacted with FZD5 to activate the WNT signaling in ESCC cells. The rW treatment also enhanced tumorigenesis and metastasis of xenograft tumors in nude mice, with these effects diminished by AW or AF treatment. This study suggests that CAFs promote growth and dissemination of ESCC cell primarily through the secretion of WNT5A.
Collapse
Affiliation(s)
- Lishuai Yao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510620, Guangdong, China
| | - Changshuai Zhou
- Department of Cardiothoracic Surgery, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
| | - Libao Liu
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510620, Guangdong, China
| | - Jinyuan He
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510620, Guangdong, China
| | - Youbo Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - An Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
14
|
Zhang Y, Liu YJ, Mei J, Yang ZX, Qian XP, Huang W. An Analysis Regarding the Association Between DAZ Interacting Zinc Finger Protein 1 (DZIP1) and Colorectal Cancer (CRC). Mol Biotechnol 2025; 67:527-547. [PMID: 38334905 DOI: 10.1007/s12033-024-01065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/21/2023] [Indexed: 02/10/2024]
Abstract
Colorectal cancer (CRC) is the third most common malignant disease worldwide, and its incidence is increasing, but the molecular mechanisms of this disease are highly heterogeneous and still far from being fully understood. Increasing evidence suggests that fibrosis mediated by abnormal activation of fibroblasts based in the microenvironment is associated with a poor prognosis. However, the function and pathogenic mechanisms of fibroblasts in CRC remain unclear. Here, combining scrna-seq and clinical specimen data, DAZ Interacting Protein 1 (DZIP1) was found to be expressed on fibroblasts and cancer cells and positively correlated with stromal deposition. Importantly, pseudotime-series analysis showed that DZIP1 levels were up-regulated in malignant transformation of fibroblasts and experimentally confirmed that DZIP1 modulates activation of fibroblasts and promotes epithelial-mesenchymal transition (EMT) in tumor cells. Further studies showed that DZIP1 expressed by tumor cells also has a driving effect on EMT and contributes to the recruitment of more fibroblasts. A similar phenomenon was observed in xenografted nude mice. And it was confirmed in xenograft mice that downregulation of DZIP1 expression significantly delayed tumor formation and reduced tumor size in CRC cells. Taken together, our findings suggested that DZIP1 was a regulator of the CRC mesenchymal phenotype. The revelation of targeting DZIP1 provides a new avenue for CRC therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
- Department of Oncology, Nanjing Tianyinshan Hospital, Nanjing, 211199, Jiangsu, China
| | - Yuan-Jie Liu
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jia Mei
- Department of Pathology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Zhao-Xu Yang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Xiao-Ping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Clinical Cancer Institute of Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Wei Huang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Hanzhong Road No.155, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
15
|
Branco H, Xavier CPR, Riganti C, Vasconcelos MH. Hypoxia as a critical player in extracellular vesicles-mediated intercellular communication between tumor cells and their surrounding microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189244. [PMID: 39672279 DOI: 10.1016/j.bbcan.2024.189244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
In the past years, increasing attention has been paid to the role of extracellular vesicles (EVs) as mediators of intercellular communication in cancer. These small size particles mediate the intercellular transfer of important bioactive molecules involved in malignant initiation and progression. Hypoxia, or low partial pressure of oxygen, is recognized as a remarkable feature of solid tumors and has been demonstrated to exert a profound impact on tumor prognosis and therapeutic efficacy. Indeed, the high-pitched growth rate and chaotic neovascular architecture that embodies solid tumors results in a profound reduction in oxygen pressure within the tumor microenvironment (TME). In response to oxygen-deprived conditions, tumor cells and their surrounding milieu develop homeostatic adaptation mechanisms that contribute to the establishment of a pro-tumoral phenotype. Latest evidence suggests that the hypoxic microenvironment that surrounds the tumor bulk may be a clincher for the observed elevated levels of circulating EVs in cancer patients. Thus, it is proposed that EVs may play a role in mediating intercellular communication in response to hypoxic conditions. This review focuses on the EVs-mediated crosstalk that is established between tumor cells and their surrounding immune, endothelial, and stromal cell populations, within the hypoxic TME.
Collapse
Affiliation(s)
- Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, 4585-116 Gandra, Portugal.
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy; Interdepartmental Research Center for Molecular Biotechnology "G. Tarone", University of Torino, 10126 Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
16
|
Jarmoshti J, Siddique A, Rane A, Mirhosseini S, Adair SJ, Bauer TW, Caselli F, Swami NS. Neural Network-Enabled Multiparametric Impedance Signal Templating for High throughput Single-Cell Deformability Cytometry Under Viscoelastic Extensional Flows. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407212. [PMID: 39439143 PMCID: PMC11798358 DOI: 10.1002/smll.202407212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Cellular biophysical metrics exhibit systematic alterations during processes, such as metastasis and immune cell activation, which can be used to identify and separate live cell subpopulations for targeting drug screening. Image-based biophysical cytometry under extensional flows can accurately quantify cell deformability based on cell shape alterations but needs extensive image reconstruction, which limits its inline utilization to activate cell sorting. Impedance cytometry can measure these cell shape alterations based on electric field screening, while its frequency response offers functional information on cell viability and interior structure, which are difficult to discern by imaging. Furthermore, 1-D temporal impedance signal trains exhibit characteristic shapes that can be rapidly templated in near real-time to extract single-cell biophysical metrics to activate sorting. We present a multilayer perceptron neural network signal templating approach that utilizes raw impedance signals from cells under extensional flow, alongside its training with image metrics from corresponding cells to derive net electrical anisotropy metrics that quantify cell deformability over wide anisotropy ranges and with minimal errors from cell size distributions. Deformability and electrical physiology metrics are applied in conjunction on the same cell for multiparametric classification of live pancreatic cancer cells versus cancer associated fibroblasts using the support vector machine model.
Collapse
Affiliation(s)
- Javad Jarmoshti
- Electrical & Computer EngineeringUniversity of VirginiaCharlottesvilleVA22904USA
| | | | - Aditya Rane
- Chemistry, University of VirginiaUniversity of VirginiaCharlottesvilleVA22904USA
| | | | - Sara J. Adair
- Surgery, School of MedicineUniversity of VirginiaCharlottesvilleVA22903USA
| | - Todd W. Bauer
- Surgery, School of MedicineUniversity of VirginiaCharlottesvilleVA22903USA
| | - Federica Caselli
- Civil Engineering and Computer ScienceUniversity of Rome Tor VergataRome00133Italy
| | - Nathan S. Swami
- Electrical & Computer EngineeringUniversity of VirginiaCharlottesvilleVA22904USA
- Chemistry, University of VirginiaUniversity of VirginiaCharlottesvilleVA22904USA
| |
Collapse
|
17
|
Vega AA, Shah PP, Rouchka EC, Clem BF, Dean CR, Woodrum N, Tanwani P, Siskind LJ, Beverly LJ. E. coli Biomolecules Increase Glycolysis and Invasive Potential in Lung Adenocarcinoma. Cancers (Basel) 2025; 17:380. [PMID: 39941749 PMCID: PMC11815989 DOI: 10.3390/cancers17030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction: Recent studies have discovered that lung cancer subtypes possess distinct microbiome profiles within their tumor microenvironment. Additionally, the tumor-associated microbiome exhibits altered bacterial pathways, suggesting that certain bacterial families are more capable of facilitating tumor progression than others. We hypothesize that there exists a crosstalk between lung adenocarcinoma (LUAD) cells and bacterial cells. Methods and Materials: RNA sequencing (RNA-seq) was performed on LUAD cell lines to explore the paracrine signaling effects of bacterial biomolecules. Based on our RNA-seq data, we investigated glycolysis by measuring glucose uptake and lactate production, invasive potential through invasion assays, and epithelial-to-mesenchymal transition (EMT) markers. Since lipopolysaccharides (LPS), abundant on the cell walls of Gram-negative bacteria, can activate toll-like receptor 4 (TLR4), we inhibited TLR4 with C34 to assess its relationship with the observed phenotypic changes. To identify the bacterial biomolecules responsible for these changes, we treated the media with RNAse enzyme, charcoal or dialyzed away molecules larger than 3 kDa. Results and Discussion: RNA-seq revealed 948 genes upregulated in the presence of E. coli biomolecules. Among these, we observed increased expression of Hexokinase II (HKII), JUN proto-oncogene, and Snail Family Transcriptional Repressor 1. We verified the elevation of glycolytic enzymes through Western blot and saw elevation of 2-deoxyglucose uptake and lactate production in LUAD cell lines incubated in E. coli biomolecules. In addition to E. coli elevating glycolysis in LUAD cell lines, E. coli exposure enhanced invasive potential as demonstrated by Boyden chamber assays. Notably, inhibition of TLR4 did not reduce the impact of E. coli biomolecules on glycolysis or the invasive potential of LUAD. Modulating the E. coli-supplemented media with RNAse enzyme or dextran-coated charcoal or using a spin column to remove biomolecules smaller than 3 kDa resulted in changes in HKII and Claudin protein expression. These findings suggest a direct relationship between E. coli and LUAD, wherein several cancer hallmarks are upregulated. Future studies should further investigate these bacterial biomolecules and their role in the tumor microenvironment to fully understand the impact of microbial shifts on cancer progression.
Collapse
Affiliation(s)
- Alexis A. Vega
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA (E.C.R.); (B.F.C.)
| | - Parag P. Shah
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Eric C. Rouchka
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA (E.C.R.); (B.F.C.)
- KY INBRE Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Brian F. Clem
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA (E.C.R.); (B.F.C.)
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Calista R. Dean
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Natassja Woodrum
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Preeti Tanwani
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Leah J. Siskind
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Levi J. Beverly
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| |
Collapse
|
18
|
Liu K, Li Y, Shen M, Xu W, Wu S, Yang X, Zhang B, Lin N. Epigenetic Regulation of Stromal and Immune Cells and Therapeutic Targets in the Tumor Microenvironment. Biomolecules 2025; 15:71. [PMID: 39858465 PMCID: PMC11764280 DOI: 10.3390/biom15010071] [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: 11/18/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The tumor microenvironment (TME) plays a pivotal role in neoplastic initiation and progression. Epigenetic machinery, governing the expression of core oncogenes and tumor suppressor genes in transformed cells, significantly contributes to tumor development at both primary and distant sites. Recent studies have illuminated how epigenetic mechanisms integrate external cues and downstream signals, altering the phenotype of stromal cells and immune cells. This remolds the area surrounding tumor cells, ultimately fostering an immunosuppressive microenvironment. Therefore, correcting the TME by targeting the epigenetic modifications holds substantial promise for cancer treatment. This review synthesizes recent research that elucidates the impact of specific epigenetic regulations-ranging from DNA methylation to histone modifications and chromatin remodeling-on stromal and immune cells within the TME. Notably, we highlight their functional roles in either promoting or restricting tumor progression. We also discuss the potential applications of epigenetic agents for cancer treatment, envisaging their ability to normalize the ecosystem. This review aims to assist researchers in understanding the dynamic interplay between epigenetics and the TME, paving the way for better epigenetic therapy.
Collapse
Affiliation(s)
- Kang Liu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Yue Li
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Minmin Shen
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Drug Clinical Trial Institution, Huzhou Central Hospital, Huzhou 313000, China
| | - Wei Xu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Shanshan Wu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Xinxin Yang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Bo Zhang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Nengming Lin
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Westlake University, Hangzhou 310024, China
| |
Collapse
|
19
|
Wang D, Li L, Zhang Y, Ye K. Lipopolysaccharide-Educated Cancer-Associated Fibroblasts Facilitate Malignant Progression of Ovarian Cancer Cells via the NF-kB/IL-6/JAK2 Signal Transduction. Mol Biotechnol 2025; 67:317-328. [PMID: 38305842 DOI: 10.1007/s12033-024-01055-3] [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: 09/08/2023] [Accepted: 12/27/2023] [Indexed: 02/03/2024]
Abstract
Gram-negative bacteria increase in ovarian cancer (OC) tissues, but its association with OC progression remains largely unknown. The present study aimed to investigate whether and how cancer-associated fibroblasts (CAFs) pretreated by the main components of bacterial outer membrane lipopolysaccharide (LPS) influence the malignancy of OC cells. Specifically, the culture medium of LPS-preconditioned CAFs (LPS-CM) further accelerated cell proliferation, colony formation and tumorigenesis of OC cells SKOV3 and HEY A8, compared with culture medium of CAFs. Next, we found that LPS pretreatment activated the nuclear factor-kappa B (NF-kB) pathway in CAFs to secret cytokines, including interleukin 1β (IL-1β), interleukin 6 (IL-6), vascular endothelial growth factor (VEGF), etc. Neutralization of IL-6 in LPS-CM abolished the promoting effect of LPS-CM on cell proliferation, survival and epithelial-mesenchymal transition (EMT) in SKOV3 and HEY A8 cells. Mechanistically, LPS-CM activated the Janus kinases 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway, while application with JAK2 inhibitor also reversed the promoting effect of LPS-CM on malignancy of OC cells. In summary, LPS-pretreated CAFs IL-6-dependently accelerate OC progression via activating the JAK2/STAT3 signal pathway, which enriches our understanding of the molecular mechanisms underlying ovaries-colonized gram-negative bacteria in OC progression.
Collapse
Affiliation(s)
- Dongjie Wang
- Department of Gynecology, The First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Lingchuan Li
- Department of Gynecology, The First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Yifeng Zhang
- Department of Gynecology, The First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, 650032, China.
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
| | - Kefan Ye
- Department of Gynecology, The First People's Hospital of Yunnan Province, No. 157, Jinbi Road, Xishan District, Kunming, 650032, China.
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
| |
Collapse
|
20
|
Yun H, Dong F, Wei X, Yan X, Zhang R, Zhang X, Wang Y. Role and value of the tumor microenvironment in the progression and treatment resistance of gastric cancer (Review). Oncol Rep 2025; 53:14. [PMID: 39611496 PMCID: PMC11622107 DOI: 10.3892/or.2024.8847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/30/2024] Open
Abstract
Gastric cancer (GC) is characterized by a complex and heterogeneous tumor microenvironment (TME) that significantly influences disease progression and treatment outcomes. The tumor stroma, which is composed of a variety of cell types such as cancer‑associated fibroblasts, immune cells and vascular components, displays significant spatial and temporal diversity. These stromal elements engage in dynamic crosstalk with cancer cells, shaping their proliferative, invasive and metastatic potential. Furthermore, the TME is instrumental in facilitating resistance to traditional chemotherapy, specific treatments and immunotherapy strategies. Understanding the underlying mechanisms by which the GC microenvironment evolves and supports tumor growth and therapeutic resistance is critical for developing effective treatment strategies. The present review explores the latest progress in understanding the intricate interactions between cancer cells and their immediate environment in GC, highlighting the implications for disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Heng Yun
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Fangde Dong
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiaoqin Wei
- Department of Pain, The Second People's Hospital of Baiyin, Baiyin, Gansu 730900, P.R. China
| | - Xinyong Yan
- Department of Proctology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Ronglong Zhang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiuyu Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Yulin Wang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| |
Collapse
|
21
|
Yang C, Shu J, Li Y, Zhao N, Liu X, Tian X, Sun Z, Tabish MS, Hong Y, Chen K, Sun M. Long non-coding RNAs are involved in the crosstalk between cancer-associated fibroblasts and tumor cells. Front Immunol 2024; 15:1469918. [PMID: 39717771 PMCID: PMC11663902 DOI: 10.3389/fimmu.2024.1469918] [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: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
The proliferation of tumors is not merely self-regulated by the cancer cells but is also intrinsically connected to the tumor microenvironment (TME). Within this complex TME, cancer-associated fibroblasts (CAFs) are pivotal in the modulation of tumor onset and progression. Rich signaling interactions exist between CAFs and tumor cells, which are crucial for tumor regulation. Long non-coding RNAs (LncRNAs) emerge from cellular transcription as a class of functionally diverse RNA molecules. Recent studies have revealed that LncRNAs are integral to the crosstalk between CAFs and tumor cells, with the capacity to modify cellular transcriptional activity and secretion profiles, thus facilitating CAFs activation, tumor proliferation, metastasis, drug resistance, and other related functionalities. This comprehensive review revisits the latest research on LncRNA-mediated interactions between CAFs and tumor cells, encapsulates the biological roles of LncRNAs, and delves into the molecular pathways from a broader perspective, aspiring to offer novel perspectives for a deeper comprehension of the etiology of tumors and the enhancement of therapeutic approaches.
Collapse
Affiliation(s)
- Chenbo Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Jiao Shu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yiwei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Zhao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiaonan Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiangyu Tian
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Zexin Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Muhammad Saud Tabish
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yichen Hong
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Asadi M, Zafari V, Sadeghi-Mohammadi S, Shanehbandi D, Mert U, Soleimani Z, Caner A, Zarredar H. The role of tumor microenvironment and self-organization in cancer progression: Key insights for therapeutic development. BIOIMPACTS : BI 2024; 15:30713. [PMID: 40256216 PMCID: PMC12008505 DOI: 10.34172/bi.30713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/08/2024] [Accepted: 11/20/2024] [Indexed: 04/22/2025]
Abstract
Introduction The tumor microenvironment (TME) plays a pivotal role in cancer progression, influencing tumor initiation, growth, invasion, metastasis, and response to therapies. This study explores the dynamic interactions within the TME, particularly focusing on self-organization-a process by which tumor cells and their microenvironment reciprocally shape one another, leading to cancer progression and resistance. Understanding these interactions can reveal new prognostic markers and therapeutic targets within the TME, such as extracellular matrix (ECM) components, immune cells, and cytokine signaling pathways. Methods A comprehensive search method was employed to investigate the current academic literature on TME, particularly focusing on self-organization in the context of cancer progression and resistance across the PubMed, Google Scholar, and Science Direct databases. Results Recent studies suggest that therapies that disrupt TME self-organization could improve patient outcomes by defeating drug resistance and increasing the effectiveness of conventional therapy. Additionally, this research highlights the essential of understanding the biophysical properties of the TME, like cytoskeletal alterations, in the development of more effective malignancy therapy. Conclusion This review indicated that targeting the ECM and immune cells within the TME can improve therapy effectiveness. Also, by focusing on TME self-organization, we can recognize new therapeutic plans to defeat drug resistance.
Collapse
Affiliation(s)
- Milad Asadi
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Venus Zafari
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Sanam Sadeghi-Mohammadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ufuk Mert
- Institute of Health Sciences, Department of Basic Oncology, Ege University, Izmir, Turkey
| | - Zahra Soleimani
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayşe Caner
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Yang C, Liu W, Powell CA, Wang Q. Heterogeneity and therapeutic implications of cancer-associated fibroblasts in lung cancer: Recent advances and future perspectives. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:240-249. [PMID: 39834587 PMCID: PMC11742357 DOI: 10.1016/j.pccm.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Indexed: 01/22/2025]
Abstract
Lung cancer is a leading cause of cancer-related mortality. The tumor microenvironment is a complex and heterogeneous cellular environment surrounding tumor cells, including cancer-associated fibroblasts (CAFs), blood vessels, immune cells, the extracellular matrix, and various cytokines secreted by cells. CAFs are highly heterogeneous and play crucial roles in lung cancer. This review highlights recent advances in the understanding of CAFs in lung cancer, focusing on their heterogeneity and functions in tumorigenesis, progression, angiogenesis, invasion, metastasis, therapy resistance, tumor immune suppression, and targeted therapy responses. Additionally, we explore the underlying mechanisms and the potential of CAFs as a target in the development of innovative therapies for lung cancer.
Collapse
Affiliation(s)
- Chunhui Yang
- Department of Clinical Laboratory, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| | - Wenwen Liu
- Translational Research Center for Lung Cancer, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| | - Charles A. Powell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Qi Wang
- Translational Research Center for Lung Cancer, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| |
Collapse
|
24
|
Wu F, Xu Y. Immunogenic cell death-related cancer-associated fibroblast clusters and prognostic risk model in cervical cancer. APL Bioeng 2024; 8:046114. [PMID: 39691350 PMCID: PMC11650426 DOI: 10.1063/5.0240772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/01/2024] [Indexed: 12/19/2024] Open
Abstract
Cervical cancer (CC) remains a leading cause of female cancer mortality globally. Immunogenic cell death (ICD) influences the tumor microenvironment (TME) and adaptive immune responses. Cancer-associated fibroblasts (CAFs) within the TME suppress anti-tumor immunity and contribute to CC progression. This study identified three ICD-related CAF clusters linked to patient survival, including IL6+CAF and ILR1+CAF, which were associated with clinical outcomes. Using a nine-gene risk model, patients were stratified into risk groups, with high-risk individuals showing worse survival and correlations with pathways such as hypoxia and TGFβ. The model also predicted immunotherapy responses, highlighting immune infiltration differences across risk groups. These findings provide insights into the role of CAF clusters in CC and present a risk model that supports prognosis prediction and personalized therapy.
Collapse
Affiliation(s)
- Fei Wu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Yue Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| |
Collapse
|
25
|
Liu Y, Pan J, Jing F, Chen X, Zhao X, Zhang J, Zhang Z, Wang J, Dai M, Wang N, Zhao X, Han J, Wang T, Chen X, Yuan H. Comparison of 68Ga-FAPI-04 and 18F-FDG PET/CT in diagnosing ovarian cancer. Abdom Radiol (NY) 2024; 49:4531-4542. [PMID: 38937339 DOI: 10.1007/s00261-024-04469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE This study assesses the diagnostic performance of 68Ga-FAPI-04 PET/CT compared to 18F-FDG PET/CT in primary, recurrent, and metastatic ovarian cancer. METHODS Seventy-nine ovarian cancer patients who performed 68Ga-FAPI-04 and 18F-FDG PET/CT were recruited. The target-to-background ratio (TBR), maximum standardized uptake value (SUVmax), the number of positive lesions, visual assessment, the peritoneal cancer index (PCI) score, staging/restaging, and treatment strategies were compared from the corresponding PET/CT. Additionally, we analyzed and contrasted the diagnostic efficacy in both scans. RESULTS Among all patients, 6 were assessed for initial assessment and 73 for recurrence and metastasis detection. For all lesions, 68Ga-FAPI-04 PET/CT demonstrated greater TBR than 18F-FDG PET/CT. 68Ga-FAPI-04 PET/CT demonstrated higher sensitivity for peritoneal metastases including patient-based and lesion-based analysis (95.00% vs. 83.33%, P = 0.065; 90.16% vs. 60.66%, P < 0.001) and a higher PCI score [median PCI: 6 (4, 12) vs. 4 (2, 8), P < 0.001]. According to the visual assessment, 68Ga-FAPI-04 PET revealed larger extent metastases in 55.93% (33/59) of the patients with peritoneal metastases. 68Ga-FAPI-04 was upstaged in 7 patients (8.86%, 7/79) and discrepancies in both scans caused treatment strategies to change in 11 patients (13.92%, 11/79). CONCLUSION 68Ga-FAPI-04 PET/CT outperforms 18F-FDG PET/CT in identifying metastases and can be a potential supplement for managing ovarian cancer patients.
Collapse
Affiliation(s)
- Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jiangyang Pan
- Department of Radiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Fenglian Jing
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, Hebei, China.
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Na Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Xiujuan Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Tingting Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Xiaoshan Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Huiqing Yuan
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
26
|
Ouyang Y, Wu Z, Aili D, Yang C, Zhang H, Wu T. Pan-cancer analysis identifies the oncogenic role of CCNE1 in human cancers. Aging (Albany NY) 2024; 16:13392-13408. [PMID: 39591374 PMCID: PMC11719100 DOI: 10.18632/aging.206163] [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: 03/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVE To investigate expression, prognosis, immune cell infiltration of Cyclin E1 (CCNE1) in cancer. METHODS We used TIMER and GEPIA datasets to analyze the differential expression of CCNE1 in multiple tumors. GEPIA and Kaplan-Meier plotter databases were utilized to observe the prognostic significance of CCNE1 in cancer. TIMER and cBioPortal databases were adopted for the analysis regarding immune infiltration and mutation respectively. RESULTS The results showed that CCNE1 was highly expressed in multiple cancers including BLCA, BRCA, CHOL, COAD, ESCA, HNSC, KICH, KIRC, KIRP, LIHC, LUAD, LUSC, READ, STAD, THCA, UCEC (P < 0.001) and CESC (P < 0.01). High CCNE1 expression was associated with a poor overall survival prognosis in several cancers, including ACC, BRCA, KIRC, KIRP, LGG, LIHC, LUAD and MESO. Additionally, CCNE1 expression was correlated with the cancer-associated immune infiltration level in BRCA, COAD, LUSC, STAD and THYM. CONCLUSIONS CCNE1 is expected to be a potential biomarker for tumor prognosis and immune infiltration in various cancers.
Collapse
Affiliation(s)
- Yujie Ouyang
- Department of Dermatology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Ziyi Wu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Dilihumaer Aili
- Department of Orthopedic Surgery, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi 830054, China
| | - Chunhua Yang
- Department of Orthopedics, Changsha Hospital, Xiangya Medical College, Central South University, Changsha 410005, China
| | - Hui Zhang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Tong Wu
- Department of Orthopedics, Changsha Hospital, Xiangya Medical College, Central South University, Changsha 410005, China
| |
Collapse
|
27
|
Li T, Liu Y, Dai M, Zhao X, Han J, Zhang Z, Jing F, Tian W, Zhang J, Zhao X, Wang J, Hao T, Wang T. Value of Semi-Quantitative Parameters of 68Ga-FAPI-04 PET/CT in Primary Malignant and Benign Diseases: A Comparison with 18F-FDG. Cancer Biother Radiopharm 2024; 39:654-663. [PMID: 38808470 DOI: 10.1089/cbr.2024.0026] [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: 05/30/2024] Open
Abstract
Objectives: We compared the value of the semiquantitative parameters of 68Ga-labeled FAP inhibitor (68Ga-FAPI)-04 positron emission tomography/computed tomography (PET/CT) and 18F-fluorodeoxyglucose (18F-FDG) in diagnosing primary malignant and benign diseases. Materials and Methods: 18F-FDG and 68Ga-FAPI-04 PET/CT images of 80 patients were compared. Semiquantitative parameters, including maximum standardized uptake value (SUVmax), mean SUV (SUVmean), peak SUV (SUVpeak), peak SUV by lean body mass (SULpeak), metabolic tumor volume (or tumor volume of FAPI; FAPI-TV), and TLG (or total lesion activity of FAPI; FAPI-TLA), were automatically obtained using the IntelliSpace Portal image processing workstation with a threshold of 40% SUVmax. The liver blood pool was measured as the background, and the tumor-to-background ratio (TBRliver) was calculated. Results: In all malignant lesions, FAPI-TV and FAPI-TLA were higher in 68Ga-FAPI-04 PET/CT than in 18F-FDG. In the subgroup analysis, 68Ga-FAPI-04 had higher FAPI-TV and FAPI-TLA and lower SUVmax than 18F-FDG had in group A, including gynecological tumor, esophageal, and colorectal cancers. However, six semiquantitative parameters were higher in group B (the other malignant tumors). For the benign diseases, SUVmax, SUVmean, SUVpeak, and SULpeak were lower in 68Ga-FAPI-04 PET/CT than in 18F-FDG. 68Ga-FAPI-04 PET/CT showed a lower liver background and a higher TBRliver than 18F-FDG did. 68Ga-FAPI-04 PET/CT had higher accuracy, sensitivity, and specificity than 18F-FDG had. Conclusion: More accurate semiquantitative parameters and lower abdominal background in 68Ga-FAPI-04 PET/CT make it more competitive in the differential diagnosis of malignant and benign diseases than in 18F-FDG.
Collapse
Affiliation(s)
- Tianyue Li
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
| | - Yunuan Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Meng Dai
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiujuan Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fenglian Jing
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Weiwei Tian
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tiancheng Hao
- Department of Nuclear Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tingting Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
28
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [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: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
29
|
Miripour ZS, Aminifar M, Hoseinpour P, Abbasvandi F, Karimi K, Ghahremani A, Parniani M, Ghaderinia M, Makiyan F, Aghaee P, Akbari ME, Abdolahad M. The presence of cancer-associated fibroblast in breast cavity side margins is in correlation with the expression of oncoproteins by adjacent epithelial cells: a new era in cancerous potential. J Cancer Res Clin Oncol 2024; 150:421. [PMID: 39287633 PMCID: PMC11408549 DOI: 10.1007/s00432-024-05943-8] [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: 06/29/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE Cancer-associated fibroblasts (CAFs) are one of the most critical cells in the tumor environment, with crucial roles in cancer progression and metastasis. Due to Field-Effect phenomena (also called field cancerization), the adjacent cavity side area of the margin is histologically normal, but it has been entered into neoplastic transformation due to MCT4 and MCT1 pathways activated by H2O2/ROS oxidative stress agents secreted by CAF in adjacent tumor bed microenvironment. This paper specifically focused on the role of cancer-associated fibroblast in breast tumor beds and its correlation with the presence of scattered cancer cells or onco-protein-activated cells (may be high risk but not completely transformed cancer cells) in the cavity side margins. METHODS In this study, the glycolytic behavior of non-tumoral cavity side margins was examined using carbon nanotube-based electrochemical biosensors integrated into a cancer diagnostic probe. This method enabled the detection of CAF accumulation sites in non-cancerous neighboring tissues of tumors, with a correlation to CAF concentration. Subsequently, RT-PCR, fluorescent, histopathological, and invasion assays were conducted on hyperglycolytic lesions to explore any correlation between the abundance of CAFs and the electrochemical responses of the non-cancerous tissues surrounding the tumor, as well as their neoplastic potential. RESULTS We observed overexpression of cancer-associated transcriptomes as well as the presence and hyperactivation of CAFs in cavity-side regions in which glycolytic metabolism was recorded, independent of the histopathological state of the lesion. At mean 70.4%, 66.7%, 70.4%, and 44.5% increments were observed in GLUT-1, MMP-2, N-cadherin, and MMP-9 transcriptomes by highly glycolytic but histologically cancer-free expression samples in comparison with negative controls (histologically non-cancer lesions with low glycolytic behavior). CONCLUSION The presence of CAFs is correlated with the presence of high glycolytic metabolism in the cavity margin lesion, high ROS level in the lesion, and finally aggressive cancer-associated proteins (such as MMP2, …) in the margin while these metabolomes, molecules, and proteins are absent in the margins with negatively scored CDP response and low ROS level. So, it seems that when we observe CAFs in glycolytic lesions with high ROS levels, some high-risk epithelial breast cells may exist while no histological trace of cancer cells was observed. Further research on CAFs could provide valuable insights into the local recurrence of malignant breast diseases. Hence, real-time sensors can be used to detect and investigate CAFs in the non-tumoral regions surrounding tumors in cancer patients, potentially aiding in the prevention of cancer recurrence.
Collapse
Affiliation(s)
- Zohreh Sadat Miripour
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
- UT&TUMS Cancer Electronics Research Center, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Mina Aminifar
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Parisa Hoseinpour
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
- SEPAS Pathology Lab, P. O. Box 1991945391, Tehran, Iran
| | - Fereshteh Abbasvandi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. Box 15179/64311, Tehran, Iran
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box 15179/64311, Tehran, Iran
| | - Koosha Karimi
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Alireza Ghahremani
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Mohammad Parniani
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. Box 15179/64311, Tehran, Iran
| | - Mohammadreza Ghaderinia
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Faride Makiyan
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Parisa Aghaee
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran
| | - Mohammad Esmaeil Akbari
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box 15179/64311, Tehran, Iran
| | - Mohammad Abdolahad
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, P.O. Box 14395/515, Tehran, Iran.
- UT&TUMS Cancer Electronics Research Center, University of Tehran, P.O. Box 14395/515, Tehran, Iran.
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, P.O. Box 1419733141, Tehran, Iran.
| |
Collapse
|
30
|
Ke F, Zhang R, Chen R, Guo X, Song C, Gao X, Zeng F, Liu Q. The role of Rhizoma Paridis saponins on anti-cancer: The potential mechanism and molecular targets. Heliyon 2024; 10:e37323. [PMID: 39296108 PMCID: PMC11407946 DOI: 10.1016/j.heliyon.2024.e37323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/07/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer is a disease characterized by uncontrolled cell proliferation, leading to excessive growth and invasion that can spread to other parts of the body. Traditional Chinese medicine has made new advancements in the treatment of cancer, providing new perspectives and directions for cancer treatment. Rhizoma Paridis is a widely used Chinese herbal medicine with documented anti-cancer effects dating back to ancient times. Modern research has shown that Rhizoma Paridis saponins (RPS) have various pharmacological activities. RPS can inhibit cancer in multiple ways, such as suppressing tumor growth, inducing cell cycle arrest, promoting cell apoptosis, enhancing cell autophagy, inducing ferroptosis, reducing inflammation, inhibiting angiogenesis, as well as inhibiting metastasis and invasion, and these findings demonstrate the potent anti-cancer activity of RPS. Polyphyllin I, polyphyllin II, polyphyllin VI, and polyphyllin VII have been widely reported as the main active ingredients with anti-cancer properties. Polyphyllin D, polyphyllin E, and polyphyllin G have also been confirmed to possess strong anti-cancer activity in recent years. Therefore, this review dives deep into the molecular mechanisms underlying the anti-cancer effects of RPS to serve as a valuable reference for future scientific research and their potential applications in cancer treatment.
Collapse
Affiliation(s)
- Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Can Song
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaowei Gao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
31
|
Secchia S, Beilinson V, Chen X, Yang ZF, Wayman JA, Dhaliwal J, Jurickova I, Angerman E, Denson LA, Miraldi ER, Weirauch MT, Ikegami K. Nutrient starvation activates ECM remodeling gene enhancers associated with inflammatory bowel disease risk in fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611754. [PMID: 39314475 PMCID: PMC11418948 DOI: 10.1101/2024.09.06.611754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Nutrient deprivation induces a reversible cell cycle arrest state termed quiescence, which often accompanies transcriptional silencing and chromatin compaction. Paradoxically, nutrient deprivation is associated with activated fibroblast states in pathological microenvironments in which fibroblasts drive extracellular matrix (ECM) remodeling to alter tissue environments. The relationship between nutrient deprivation and fibroblast activation remains unclear. Here, we report that serum deprivation extensively activates transcription of ECM remodeling genes in cultured fibroblasts, despite the induction of quiescence. Starvation-induced transcriptional activation accompanied large-scale histone acetylation of putative distal enhancers, but not promoters. The starvation-activated putative enhancers were enriched for non-coding genetic risk variants associated with inflammatory bowel disease (IBD), suggesting that the starvation-activated gene regulatory network may contribute to fibroblast activation in IBD. Indeed, the starvation-activated gene PLAU, encoding uPA serine protease for plasminogen and ECM, was upregulated in inflammatory fibroblasts in the intestines of IBD patients. Furthermore, the starvation-activated putative enhancer at PLAU, which harbors an IBD risk variant, gained chromatin accessibility in IBD patient fibroblasts. This study implicates nutrient deprivation in transcriptional activation of ECM remodeling genes in fibroblasts and suggests nutrient deprivation as a potential mechanism for pathological fibroblast activation in IBD.
Collapse
Affiliation(s)
- Stefano Secchia
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, USA
- Department of Biology, Lund University, Lund, 22362, Sweden
- Present address: Institute of Human Biology, Basel, Switzerland
| | - Vera Beilinson
- Department of Pediatrics, The University of Chicago, Chicago, Illinois, USA
- Present address: California Institute of Technology, Pasadena, California, USA
| | - Xiaoting Chen
- Division of Allergy and Immunology, CCHMC Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Zi F Yang
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joseph A Wayman
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jasbir Dhaliwal
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ingrid Jurickova
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Elizabeth Angerman
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily R Miraldi
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Matthew T Weirauch
- Division of Allergy and Immunology, CCHMC Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kohta Ikegami
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
32
|
Cao W, Xie Y, Cai L, Wang M, Chen Z, Wang Z, Xv J, Wang Y, Li R, Liu X, Wang W. Pan‑cancer analysis on the role of KMT2C expression in tumor progression and immunotherapy. Oncol Lett 2024; 28:444. [PMID: 39091583 PMCID: PMC11292467 DOI: 10.3892/ol.2024.14577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/10/2024] [Indexed: 08/04/2024] Open
Abstract
Histone lysine N-methyltransferase 2C (KMT2C) is involved in transcriptional regulation and DNA damage repair. Mutations in KMT2C have been implicated in the progression, metastasis, and drug resistance of multiple cancer types. However, the roles of KMT2C in the regulation of tumor prognosis, immune cell infiltration and the immune microenvironment in these multiple cancer types remain unclear. Therefore, in the present study, data from The Cancer Genome Atlas and Genotype-Tissue Expression databases were used for KMT2C expression analyses. Kaplan-Meier and univariate Cox regression analyses were also performed to investigate the prognostic role of KMT2C. In addition, Gene Set Enrichment Analysis (GSEA) was conducted to study the KMT2C-related signaling pathways. Tumor immune estimation resource 2 and single-sample GSEA were conducted to investigate the correlation between KMT2C expression and immune cell infiltrations, and Spearman's analysis was conducted to study the correlations among KMT2C, tumor mutational burden, microsatellite instability, immune regulators, chemokines and immune receptors. Immunohistochemistry of patient kidney tumor samples was performed to verify the correlation between KMT2C and programmed death-ligand 1 (PD-L1) expression. Finally, RNA interference, wound healing and colony formation assays were conducted to evaluate the effects of KMT2C expression on cell proliferation and metastasis. The results of the present study demonstrated that KMT2C was highly expressed in multiple cancer types, was a protective factor in kidney renal clear cell carcinoma and ovarian serous cystadenocarcinoma, and a risk factor for lung squamous cell carcinoma and uveal melanoma. In addition, KMT2C levels were negatively correlated with immune-activated pathways and the infiltration of immune cells, and positively correlated with inhibitory immune factors and tumor angiogenesis. Patients with low KMT2C expression had higher objective response rates to immunotherapy, and drug sensitivity analysis indicated that topoisomerase, histone deacetylase, DOT1-like histone H3K79 methyltransferase and G9A nuclear histone lysine methyltransferase inhibitors could potentially be used to treat tumors with high KMT2C expression levels. Finally, the KMT2C and PD-L1 expression levels were shown to be positively correlated, and KMT2C knockdown markedly promoted the proliferation and invasion capacities of A549 cells. In conclusion, the present study revealed that low KMT2C expression may be a promising biomarker for predicting the response of patients with cancer to immunotherapy. Conversely, high KMT2C expression was shown to promote tumor angiogenesis, which may contribute to the formation of the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Wei Cao
- Department of Thoracic Surgery, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yawen Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Li Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Mengqing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhuoying Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Ziteng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jiajia Xv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuqing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xuesong Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wenliang Wang
- Institute of Clinical Immunology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
33
|
Xu W, Liu S, Ma L, Cheng L, Li Q, Qing L, Yang Y, Dong Z. Identification of miRNA signature in cancer-associated fibroblast to predict recurrent prostate cancer. Comput Biol Med 2024; 180:108989. [PMID: 39142223 DOI: 10.1016/j.compbiomed.2024.108989] [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: 04/17/2024] [Revised: 07/16/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are one of the major components of prostate stromal cells, which play a crucial part in tumor development and treatment resistance. This study aimed to establish a model of CAFs-related microRNAs (miRNAs) to assess prognostic differences, tumor microenvironments, and screening of anticancer drugs by integrating data from single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (buRNA-seq). METHODS scRNA-seq and buRNA-seq data of primary prostate cancer (PCa) were downloaded from Gene Expression Omnibus and The Cancer Genome Atlas databases. Statistical methods including Least absolute shrinkage and selection operator (Lasso), Lasso penalized, Random Forest, Random Forest Combination, and Support Vector Machine (SVM) were performed to select hub miRNAs. Pathway analyses and assessment of infiltrating immune cells were conducted using Gene Set Enrichment Analysis and the CIBERSORT algorithm. The expression of CAFs-related miRNAs in fibroblast cell lines were validated through quantitative real-time PCR. Cell Counting Kit 8 (CCK8), wound-healing, clone formation, and cell migration assays were used to explore cell proliferation, growth, and migration in vitro. A mouse xenograft model was established to investigate the effect of CAFs on tumor growth in vivo. RESULTS Through single-cell transcriptomics analysis in 34 PCa patients, 89 CAFs-related mRNAs were identified. A prognostic model based on 9 CAFs-related miRNAs (hsa-miR-1258, hsa-miR-133b, hsa-miR-222-3p, hsa-miR-145-3p, hsa-miR-493-5p, hsa-miR-96-5p, hsa-miR-15b-5p, hsa-miR-106b-5p, and hsa-miR-191-5p) was established to predict biochemical recurrence (BCR). We have determined through two prediction methods that NVP-TAE684 may be the optimal targeted therapy drug for treating CAFs. Downregulation of hsa-miR-106b-5p in CAFs significantly suppressed cell proliferation, migration, and colony formation in vitro. In vivo studies using a xenograft model further confirmed that hsa-miR-106b-5p downregulation significantly reduced tumor growth. CONCLUSION Our findings conducted an integrated bioinformatic analysis to develop a CAFs-related miRNAs model that provides prognostic insights into individualized and precise treatment for prostate adenocarcinoma patients. Downregulation of miR-106b-5p in CAFs significantly suppressed tumor growth, suggesting a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xu
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| | - Shuai Liu
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| | - Longtu Ma
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| | - Long Cheng
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| | - Qingchao Li
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| | - Liangliang Qing
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| | - Yongjin Yang
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| | - Zhilong Dong
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China; Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
34
|
Farag MA, Kandeel MM, Kassab AE, Faggal SI. Medicinal attributes of thienopyrimidine scaffolds incorporating the aryl urea motif as potential anticancer candidates via VEGFR inhibition. Arch Pharm (Weinheim) 2024; 357:e2400125. [PMID: 38738795 DOI: 10.1002/ardp.202400125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024]
Abstract
Worldwide, cancer is a major public health concern. It is a well-acknowledged life-threatening disease. Despite numerous advances in the understanding of the genetic basis of cancer growth and progression, therapeutic challenges remain high. Human tumors exhibited mutation or overexpression of several tyrosine kinases (TK). The vascular endothelial growth factor receptor (VEGFR) is a TK family member and is well known for tumor growth and progression. Therefore, VEGF/VEGFR pathway inhibition is an appealing approach for cancer drug discovery. This review will discuss the structure-based optimization of thienopyrimidines incorporating the aryl urea moiety to develop scaffolds of potent anticancer activity via VEGFR inhibition published between 2013 and 2023. Increasing knowledge of probable scaffolds that can act as VEGFR inhibitors might spur the hunt for novel anticancer medications that are safer, more effective, or both.
Collapse
Affiliation(s)
- Myrna A Farag
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Manal M Kandeel
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Samar I Faggal
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
35
|
Na L, Fan F. Advances in nanobubbles for cancer theranostics: Delivery, imaging and therapy. Biochem Pharmacol 2024; 226:116341. [PMID: 38848778 DOI: 10.1016/j.bcp.2024.116341] [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: 03/07/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Maximizing treatment efficacy and forecasting patient prognosis in cancer necessitates the strategic use of targeted therapy, coupled with the prompt precise detection of malignant tumors. Theutilizationof gaseous systems as an adaptable platform for creating nanobubbles (NBs) has garnered significant attention as theranostics, which involve combining contrast chemicals typically used for imaging with pharmaceuticals to diagnose and treattumorssynergistically in apersonalizedmanner for each patient. This review specifically examines the utilization of oxygen NBsplatforms as a theranostic weapon in the field of oncology. We thoroughly examine the key factors that impact the effectiveness of NBs preparations and the consequences of these treatment methods. This review extensively examines recent advancements in composition schemes, advanced developments in pre-clinical phases, and other groundbreaking inventions in the area of NBs. Moreover, this review offers a thorough examination of the optimistic future possibilities, addressing prospective methods for improvement and incorporation into widely accepted therapeutic practices. As we explore the ever-changing field of cancer theranostics, the incorporation of oxygen NBs appears as a promising development, providing new opportunities for precision medicine and marking a revolutionary age in cancer research and therapy.
Collapse
Affiliation(s)
- Liu Na
- Ultrasound Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Fan Fan
- School of Automation, Xi'an University of Posts and Telecommunications, Xi'an 710121, China.
| |
Collapse
|
36
|
Zhang Y, Chen H, Zhang W, Zhou H. Identification of cancer-associated fibroblast-related Ectodysplasin-A as a novel indicator for prognosis and immune response in gastric cancer. Heliyon 2024; 10:e34005. [PMID: 39091933 PMCID: PMC11292546 DOI: 10.1016/j.heliyon.2024.e34005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Studies have indicated cancer-associated fibroblasts (CAFs) could have a significant impact in gastric cancer (GC) progression and chemotherapy resistance. However, the gene related to cancer fibroblasts that can be used as biomarkers to judge the occurrence of gastric cancer has not been fully explored. Based on two Gene Expression Omnibus (GEO) datasets, we focus on differentially expressed genes which may act as CAFs markers related to GC. Through COX regression, LASSO regression and Kaplan-Meier survival analysis, we discovered three upregulated genes (GLT8D2, GNAS and EDA) associated with poor GC patients' survival. By single-cell analysis and nomogram, we found that EDA may affect fibroblast production and disease prognosis in GC patients. EDA expression showed a positive correlation with 5-Fluorouracil IC50 values. Immunohistochemistry (IHC) and real time PCR indicated elevated EDA levels in GC tissues and cells. Enrichment analysis revealed that EDA was closely linked to immune system regulation. IHC and single-cell analysis indicated that EDA gene was associated with cancer fibroblasts marker FGF12 and influence cell interferon-gamma response, which may play a role in regulating immune-related characteristics. In summary, we concluded that EDA may be used as a new therapeutic CAFs marker for GC.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Pathology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Haoran Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenzheng Zhang
- Department of Joint and Sports Medicine, Taian City Central Hospital, Taian, Shandong, China
| | - Haiyan Zhou
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
37
|
Huang H, Wang Q, Tan J, Zeng C, Wang J, Huang J, Hu Y, Wu Q, Wu X, Liu C, Ye X, Fan Y, Sun W, Guo Z, Peng L, Zou L, Xiang D, Song Y, Zheng X, Wan Y. Quinoa greens as a novel plant food: a review of its nutritional composition, functional activities, and food applications. Crit Rev Food Sci Nutr 2024:1-21. [PMID: 38993144 DOI: 10.1080/10408398.2024.2370483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Quinoa (Chenopodium quinoa Willd) is widely regarded as a versatile pseudo-cereal native to the Andes Mountains in South America. It has gained global recognition as a superfood due to its rich nutritional profile. While quinoa grains are well-known, there is an undiscovered potential in quinoa greens, such as sprouts, leaves, and microgreens. These verdant parts of quinoa are rich in a diverse array of essential nutrients and bioactive compounds, including proteins, amino acids, bioactive proteins, peptides, polyphenols, and flavonoids. They have powerful antioxidant properties, combat cancer, and help prevent diabetes. Quinoa greens offer comparable or even superior benefits when compared to other sprouts and leafy greens, yet they have not gained widespread recognition. Limited research exists on the nutritional composition and biological activities of quinoa greens, underscoring the necessity for thorough systematic reviews in this field. This review paper aims to highlight the nutritional value, bioactivity, and health potential of quinoa greens, as well as explore their possibilities within the food sector. The goal is to generate interest within the research community and promote further exploration and wider utilization of quinoa greens in diets. This focus may lead to new opportunities for enhancing health and well-being through innovative dietary approaches.
Collapse
Affiliation(s)
- Huange Huang
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Qiang Wang
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Jianxin Tan
- Institute of Agriculture, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lasa, China
| | - Chunxiang Zeng
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Junying Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jingwei Huang
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Yichen Hu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Qi Wu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Xiaoyong Wu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Changying Liu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Xueling Ye
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Yu Fan
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Wenjun Sun
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Zhanbin Guo
- College of Agronomy, Inner Mongolia Agricultural University, Inner Mongolia, China
| | - Lianxin Peng
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Dabing Xiang
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Yu Song
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Xiaoqin Zheng
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Yan Wan
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| |
Collapse
|
38
|
Kaviani M, Soleimanian S, Keshtkar S, Azarpira N, Asvar Z, Pakbaz S. Molecular Prospective on Malignant Transformation of Mesenchymal Stem Cells: An Issue in Cell Therapy. Cell Reprogram 2024; 26:96-106. [PMID: 38917438 DOI: 10.1089/cell.2024.0026] [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: 06/27/2024] Open
Abstract
Mesenchymal stem cell (MSCs) therapy, as a rapidly developing area of medicine, holds great promise for the treatment of a variety of medical conditions. MSCs are multipotent stem cells that can be isolated from various tissues and could self-renew and differentiate. They secrete cytokines and trophic factors that create a regenerative microenvironment and have immunomodulatory properties. Although clinical trials have been conducted with MSCs in various diseases, concerns regarding the possibility of malignant transformation of these cells have been raised. The studies showed a higher rate of hematological malignancy and carcinogenesis in experimental models after MSC transplantation. The mechanisms underlying malignant transformation of MSCs are complex and not fully understood, but they are believed to involve the presence of special signaling molecules and alterations in cell behavior regulation pathways. Possible pathways that lead to MSCs' oncogenic transformation occur through two mechanisms: spontaneous and stimulated malignant transformation, including cell fusion, fusion proteins, and the tumor microenvironment. MSC-based therapies have the potential to revolutionize medicine, and addressing the issue of malignancy is crucial to ensure their safety and efficacy. Therefore, the purpose of the present review is to summarize the potential mechanisms of the malignant transformation of MSCs. [Figure: see text].
Collapse
Affiliation(s)
- Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Keshtkar
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Asvar
- Nanotechnology School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Pakbaz
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
39
|
Liu Y, Han X, Han Y, Bi J, Wu Y, Xiang D, Zhang Y, Bi W, Xu M, Li J. Integrated transcriptomic analysis systematically reveals the heterogeneity and molecular characterization of cancer-associated fibroblasts in osteosarcoma. Gene 2024; 907:148286. [PMID: 38367852 DOI: 10.1016/j.gene.2024.148286] [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: 11/15/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND Osteosarcoma (OS), with a peak incidence during the adolescent growth spurt, is correlated with poor prognosis for its high malignancy. The tumor microenvironment (TME) is highly complicated, with frequent interactions between tumor and stromal cells. The cancer-associated fibroblasts (CAFs) in the TME have been considered to actively involve in the progression, metastasis, and drug resistance of OS. This study aimed to characterize cellular heterogeneity and molecular characterization in CAFs subtypes and explore the potential targeting therapeutic strategies to improve the prognosis of OS patients. METHODS The single-cell atlas of human OS tumor lesions were constructed from the GEO database. Then significant marker genes and potential biological functions for each CAFs subtype were identified and explored using the Seurat R package. Next, by performing the survival analyses and constructing the risk scores for CAFs subtypes, we aimed to identify and characterize the prognostic values of specific marker genes and different CAFs subtypes. Furthermore, we explored the therapeutic targets and innovative drugs targeting different CAFs subtypes based on the GDSC database. Finally, prognoses related CAFs subtypes were further validated through immunohistochemistry (IHC) on clinical OS specimens. RESULTS Overall, nine main cell clusters and five subtypes of CAFs were identified. The differentially expressed marker genes for each CAFs clusters were then identified. Moreover, through Gene Ontology (GO) enrichment analysis, we defined the CAFs_2 (upregulated CXCL14 and C3), which was closely related to leukocyte migration and chemotaxis, as inflammatory CAFs (iCAFs). Likewise, we defined the CAFs_4 (upregulated CD74, HLA-DRA and HLA-DRB1), which was closely related to antigen process and presentation, as antigen-presenting CAFs (apCAFs). Furthermore, Kaplan-Meier analyses showed that CAFs_2 and CAFs_4 were correlated with poor clinical prognosis of OS patients. Meanwhile, therapeutic drugs targeting CAFs_2 and CAFs_4, such as 17-AAG/Docetaxel/Bleomycin and PHA-793887/NG-25/KIN001-102, were also explored, respectively. Finally, IHC assay confirmed the abundant CAFs_2 and CAFs_4 subtypes infiltration in the OS microenvironment compared with adjacent tissues. CONCLUSION Our study revealed the diversity, complexity, and heterogeneity of CAFs in OS, and complemented the single-cell atlas in OS TME.
Collapse
Affiliation(s)
- Yuyang Liu
- Department of Neurosurgery, 920th Hospital of Joint Logistics Support Force, Kunming 650032, Yunnan, China; Chinese PLA Spinal Cord Injury Treatment Center, Kunming, Yunnan 650032, China
| | - Xinli Han
- School of Medicine, Nankai University, Tianjin 300074, China
| | - Yuchen Han
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Jingyou Bi
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yanan Wu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Dongquan Xiang
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yinglong Zhang
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Wenzhi Bi
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China; School of Medicine, Nankai University, Tianjin 300074, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Meng Xu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China; Medical School of Chinese PLA, Beijing 100853, China.
| | - Jianxiong Li
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
40
|
Liu Y, Wang Y, Yu Z, Wang Z. Impacts of TP53TG1 in cancer-associated fibroblasts-derived exosomes on epithelial-mesenchymal transition capacity of colorectal carcinoma cells by targeting miR-330-3p. Heliyon 2024; 10:e30301. [PMID: 38707274 PMCID: PMC11068805 DOI: 10.1016/j.heliyon.2024.e30301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Objective This research aims at clarifying the action and mechanisms of action of TP53TG1 in cancer-associated fibroblasts (CAF)-derived exosomes (EXs) on colorectal carcinoma (CRC) cells. Methods CAF and CAF-EXs isolated from CRC tissues were incubated with CRC SW480 cells to determine alterations in biological behavior, epithelial-mesenchymal transition (EMT) capacity, and TP53TG1 and miR-330-3p expression. In addition, a dual luciferase reporter (DLR) assay was conducted to verify the connection between TP53TG1 and miR-330-3p, and the impacts of the two genes on CRC cells were analyzed. Results CRC-CAF-EXs extracted from CRC tissues were successfully identified and were able to promote SW480 multiplication, invasiveness, migration, and EMT ability while inhibiting apoptosis (P < 0.05). In addition, TP53TG1 increased and miR-330-3p decreased in SW480 when cultured with CRC-CAF-EXs (P < 0.05). The DLR assay identified notably reduced fluorescence activity of TP53TG1-WT after transfection with miR-330-3p-mimics (P < 0.05). Furthermore, SW480 cell multiplication, invasiveness and migration were found to be enhanced and the apoptosis decreased after up-regulating TP53TG1, while suppressing TP53TG1 and up-regulating miR-330-3p contributed to quite the opposite effect (P < 0.05). Moreover, by elevating TP53TG1 and miR-330-3p simultaneously, we found a cell activity similar to the NC group (P > 0.05). Conclusion By targeting miR-330-3p, TP53TG1 in CRC-CAF-EXs can enhance CRC cell activity and EMT capacity and inhibit apoptosis.
Collapse
Affiliation(s)
- Yawei Liu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Youwei Wang
- Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Zhijuan Yu
- Hubei University of Science and Technology, Xianning, Hubei, 437000, China
| | - Ziheng Wang
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
- Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, Jiangsu, 215000, China
- The School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
41
|
Bai Y, Li L, Li J, Lu X. Association analysis of FXYD5 with prognosis and immunological characteristics across pan-cancer. Heliyon 2024; 10:e30727. [PMID: 38774095 PMCID: PMC11107115 DOI: 10.1016/j.heliyon.2024.e30727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Background The FXYD domain-containing ion transport regulator 5 (FXYD5) gene is a cancer promoter. However, evidence for an association between FXYD5 and various types of cancer is still lacking. Using multi-omics bioinformatics, our study aimed to reveal the expression distribution, prognostic value, immune infiltration correlation, and molecular functions of FXYD5. Methods Using pan-cancer multi-omics data (including The Cancer Genome Atlas, PrognoScan, Gene Expression Profiling Interactive Analysis, cBioPortal, Gene Expression Omnibus, TIMER and scTIME Portal), we assessed the differences in the expression and prognostic value of FXYD5 in malignant tumors. Furthermore, at the single-cell level, we analyze the expression distribution of FXYD5 across different cell types within the tumor microenvironment, and its relationship with the immune microenvironment. Finally, focusing on ovarian cancer, we conducted preliminary validation of the above findings using cell and molecular biology techniques. Results Our results indicated that FXYD5 was up-regulated in various tumor types and was positively associated with tumor progression. We also revealed that FXYD5 was ubiquitously expressed in microenvironmental cells at the single-cell level, and its upregulation was associated with enhanced immune infiltration, cancer-associated fibroblast infiltration, and dysfunction of tumor-infiltrating cytotoxic T lymphocyte. Additionally, its expression was positively correlated with immune checkpoint genes, DNA mismatch repair genes, MSI (microsatellite instability) and TMB (tumor mutational burden) across various cancers. Its higher expression in cytotoxic T lymphocytes attenuated its ability to predict patient survival with PD-L1 (programmed death-ligand 1) blockade therapy, and FXYD5 was found to be a potential regulator of tumor immune escape and resistance to cancer immunotherapies. Based on GSEA (gene set enrichment analysis) and experimental verification, FXYD5 activated TGF-β/SMAD signaling and drove EMT (epithelial-mesenchymal transition) to promote ovarian cancer progression. Conclusion In summary, our study revealed that FXYD5-TGFβ axis may coregulate the interaction between tumors, CAFs (carcinoma-associated fibroblasts) and immune cells to reshape the tumor immune microenvironment and promote tumorigenesis and tumor progression. Thus, FXYD5 could be used as an immune-related biomarker for diagnosing and predicting the prognosis of multiple cancer types. Therefore, our findings suggest that targeting FXYD5 in TME (tumor microenvironment) may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yang Bai
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Liangdong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Xin Lu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| |
Collapse
|
42
|
Mori JO, Elhussin I, Brennen WN, Graham MK, Lotan TL, Yates CC, De Marzo AM, Denmeade SR, Yegnasubramanian S, Nelson WG, Denis GV, Platz EA, Meeker AK, Heaphy CM. Prognostic and therapeutic potential of senescent stromal fibroblasts in prostate cancer. Nat Rev Urol 2024; 21:258-273. [PMID: 37907729 PMCID: PMC11058122 DOI: 10.1038/s41585-023-00827-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
The stromal component of the tumour microenvironment in primary and metastatic prostate cancer can influence and promote disease progression. Within the prostatic stroma, fibroblasts are one of the most prevalent cell types associated with precancerous and cancerous lesions; they have a vital role in the structural composition, organization and integrity of the extracellular matrix. Fibroblasts within the tumour microenvironment can undergo cellular senescence, which is a stable arrest of cell growth and a phenomenon that is emerging as a recognized hallmark of cancer. Supporting the idea that cellular senescence has a pro-tumorigenic role, a subset of senescent cells exhibits a senescence-associated secretory phenotype (SASP), which, along with increased inflammation, can promote prostate cancer cell growth and survival. These cellular characteristics make targeting senescent cells and/or modulating SASP attractive as a potential preventive or therapeutic option for prostate cancer.
Collapse
Affiliation(s)
- Joakin O Mori
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Isra Elhussin
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mindy K Graham
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clayton C Yates
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel R Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William G Nelson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerald V Denis
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elizabeth A Platz
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alan K Meeker
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher M Heaphy
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
43
|
Janani M, Poorkhani A, Amiriani T, Donyadideh G, Ahmadi F, Jorjanisorkhankalateh Y, Beheshti-Nia F, Kalaei Z, Roudbaraki M, Soltani M, Khori V, Alizadeh AM. Association of future cancer metastases with fibroblast activation protein-α: a systematic review and meta-analysis. Front Oncol 2024; 14:1339050. [PMID: 38751814 PMCID: PMC11094201 DOI: 10.3389/fonc.2024.1339050] [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: 04/04/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Fibroblast activation protein-α (FAP-α) is a vital surface marker of cancer-associated fibroblasts, and its high expression is associated with a higher tumor grade and metastasis. A systematic review and a meta-analysis were performed to associate future metastasis with FAP-α expression in cancer. Methods In our meta-analysis, relevant studies published before 20 February 2024 were systematically searched through online databases that included PubMed, Scopus, and Web of Science. The association between FAP-α expression and metastasis, including distant metastasis, lymph node metastasis, blood vessel invasion, vascular invasion, and neural invasion, was evaluated. A pooled odds ratio (OR) with 95% confidence intervals (CI) was reported as the measure of association. Results A total of 28meta-analysis. The random-effects model for five parameters showed that a high FAP-α expression was associated with blood vessel invasion (OR: 3.04, 95% CI: 1.54-5.99, I 2 = 63%, P = 0.001), lymphovascular invasion (OR: 3.56, 95% CI: 2.14-5.93, I 2 = 0.00%, P < 0.001), lymph node metastasis (OR: 2.73, 95% CI: 1.96-3.81, I 2 = 65%, P < 0.001), and distant metastasis (OR: 2.59; 95% CI: 1.16-5.79, I 2 = 81%, P < 0.001). However, our analysis showed no statistically significant association between high FAP-α expression and neural invasion (OR: 1.57, 95% CI: 0.84-2.93, I 2 = 38%, P = 0.161). Conclusions This meta-analysis indicated that cancer cells with a high FAP-α expression have a higher risk of metastasis than those with a low FAP-α expression. These findings support the potential importance of FAP-α as a biomarker for cancer metastasis prediction.
Collapse
Affiliation(s)
- Majid Janani
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhoushang Poorkhani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ghazaleh Donyadideh
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farahnazsadat Ahmadi
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Fereshteh Beheshti-Nia
- Department of Epidemiology and Biostatistics, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Kalaei
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Morad Roudbaraki
- Laboratory of Cell Physiology, Inserm U1003, University of Lille, Villeneuve d’Ascq, France
| | - Mahsa Soltani
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Mohammad Alizadeh
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Contreras-Martínez OI, Angulo-Ortíz A, Santafé Patiño G, Rocha FV, Zanotti K, Fortaleza DB, Teixeira T, Sierra Martinez J. Cytotoxic Potential of the Monoterpene Isoespintanol against Human Tumor Cell Lines. Int J Mol Sci 2024; 25:4614. [PMID: 38731832 PMCID: PMC11083712 DOI: 10.3390/ijms25094614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer is a disease that encompasses multiple and different malignant conditions and is among the leading causes of death in the world. Therefore, the search for new pharmacotherapeutic options and potential candidates that can be used as treatments or adjuvants to control this disease is urgent. Natural products, especially those obtained from plants, have played an important role as a source of specialized metabolites with recognized pharmacological properties against cancer, therefore, they are an excellent alternative to be used. The objective of this research was to evaluate the action of the monoterpene isoespintanol (ISO) against the human tumor cell lines MDA-MB-231, A549, DU145, A2780, A2780-cis and the non-tumor line MRC-5. Experiments with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and fluorescence with propidium iodide (PI), 4',6-diamidino-2-phenylindole dilactate (DAPI) and green plasma revealed the cytotoxicity of ISO against these cells; furthermore, morphological and chromogenic studies revealed the action of ISO on cell morphology and the inhibitory capacity on reproductive viability to form colonies in MDA-MB-231 cells. Likewise, 3D experiments validated the damage in these cells caused by this monoterpene. These results serve as a basis for progress in studies of the mechanisms of action of these compounds and the development of derivatives or synthetic analogues with a better antitumor profile.
Collapse
Affiliation(s)
| | - Alberto Angulo-Ortíz
- Chemistry Department, Faculty of Basic Sciences, University of Córdoba, Montería 230002, Colombia;
| | - Gilmar Santafé Patiño
- Chemistry Department, Faculty of Basic Sciences, University of Córdoba, Montería 230002, Colombia;
| | - Fillipe Vieira Rocha
- Chemistry Department, Federal University of São Carlos, São Carlos 13565-905, Brazil; (F.V.R.); (K.Z.); (T.T.)
| | - Karine Zanotti
- Chemistry Department, Federal University of São Carlos, São Carlos 13565-905, Brazil; (F.V.R.); (K.Z.); (T.T.)
| | - Dario Batista Fortaleza
- Chemistry Department, Federal University of São Carlos, São Carlos 13565-905, Brazil; (F.V.R.); (K.Z.); (T.T.)
| | - Tamara Teixeira
- Chemistry Department, Federal University of São Carlos, São Carlos 13565-905, Brazil; (F.V.R.); (K.Z.); (T.T.)
| | - Jesus Sierra Martinez
- Genetics and Evolution Department, Federal University of São Carlos, São Carlos 13565-905, Brazil
| |
Collapse
|
45
|
Song N, Cui K, Zeng L, Li M, Fan Y, Shi P, Wang Z, Su W, Wang H. Advance in the role of chemokines/chemokine receptors in carcinogenesis: Focus on pancreatic cancer. Eur J Pharmacol 2024; 967:176357. [PMID: 38309677 DOI: 10.1016/j.ejphar.2024.176357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
The chemokines/chemokine receptors pathway significantly influences cell migration, particularly in recruiting immune cells to the tumor microenvironment (TME), impacting tumor progression and treatment outcomes. Emerging research emphasizes the involvement of chemokines in drug resistance across various tumor therapies, including immunotherapy, chemotherapy, and targeted therapy. This review focuses on the role of chemokines/chemokine receptors in pancreatic cancer (PC) development, highlighting their impact on TME remodeling, immunotherapy, and relevant signaling pathways. The unique immunosuppressive microenvironment formed by the interaction of tumor cells, stromal cells and immune cells plays an important role in the tumor proliferation, invasion, migration and therapeutic resistance. Chemokines/chemokine receptors, such as chemokine ligand (CCL) 2, CCL3, CCL5, CCL20, CCL21, C-X-C motif chemokine ligand (CXCL) 1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, CXCL14, CXCL16, CXCL17, and C-X3-C motif chemokine ligand (CX3CL)1, derived mainly from leukocyte cells, cancer-related fibroblasts (CAFs), pancreatic stellate cells (PSCs), and tumor-associated macrophages (TAMs), contribute to PC progression and treatment resistance. Chemokines recruit myeloid-derived suppressor cells (MDSC), regulatory T cells (Tregs), and M2 macrophages, inhibiting the anti-tumor activity of immune cells. Simultaneously, they enhance pathways like epithelial-mesenchymal transition (EMT), Akt serine/threonine kinase (AKT), extracellular regulated protein kinases (ERK) 1/2, and nuclear factor kappa-B (NF-κB), etc., elevating the risk of PC metastasis and compromising the efficacy of radiotherapy, chemotherapy, and anti-PD-1/PD-L1 immunotherapy. Notably, the CCLx-CCR2 and CXCLx-CXCR2/4 axis emerge as potential therapeutic targets in PC. This review integrates recent findings on chemokines and receptors in PC treatment, offering valuable insights for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Na Song
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Kai Cui
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Liqun Zeng
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Mengxiao Li
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China
| | - Yanwu Fan
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Pingyu Shi
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Ziwei Wang
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Wei Su
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China.
| | - Haijun Wang
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China.
| |
Collapse
|
46
|
Akter R, Awais M, Boopathi V, Ahn JC, Yang DC, Kang SC, Yang DU, Jung SK. Inversion of the Warburg Effect: Unraveling the Metabolic Nexus between Obesity and Cancer. ACS Pharmacol Transl Sci 2024; 7:560-569. [PMID: 38481689 PMCID: PMC10928896 DOI: 10.1021/acsptsci.3c00301] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 02/04/2025]
Abstract
Obesity is a well-established risk factor for cancer, significantly impacting both cancer incidence and mortality. However, the intricate molecular mechanisms connecting adipose tissue to cancer cell metabolism are not fully understood. This Review explores the historical context of tumor energy metabolism research, tracing its origins to Otto Warburg's pioneering work in 1920. Warburg's discovery of the "Warburg effect", wherein cancer cells prefer anaerobic glycolysis even in the presence of oxygen, laid the foundation for understanding cancer metabolism. Building upon this foundation, the "reverse Warburg effect" emerged in 2009, elucidating the role of aerobic glycolysis in cancer-associated fibroblasts (CAFs) and its contribution to lactate accumulation in the tumor microenvironment, subsequently serving as a metabolic substrate for cancer cells. In contrast, within high-adiposity contexts, cancer cells exhibit a unique metabolic shift termed the "inversion of the Warburg effect". This phenomenon, distinct from the stromal-dependent reverse Warburg effect, relies on increased nutrient abundance in obesity environments, leading to the generation of glucose from lactate as a metabolic substrate. This Review underscores the heightened tumor proliferation and aggressiveness associated with obesity, introducing the "inversion of the Warburg effect" as a novel mechanism rooted in the altered metabolic landscape within an obese milieu. The insights presented here open promising avenues for therapeutic exploration, offering fresh perspectives and opportunities for the development of innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Reshmi Akter
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Muhammad Awais
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Vinothini Boopathi
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Jong Chan Ahn
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Deok Chun Yang
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Se Chan Kang
- Graduate
School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggido, Republic of Korea
| | - Dong Uk Yang
- AIBIOME, 6, Jeonmin-ro 30beon-gil, Yuseong-gu 34214, Daejeon, Republic of Korea
| | - Seok-Kyu Jung
- Department
of Horticulture, Kongju National University, Yesan 32588,Chungcheongnam-do, Republic of Korea
| |
Collapse
|
47
|
Lin S, Zhu B. Exosome-transmitted FOSL1 from cancer-associated fibroblasts drives colorectal cancer stemness and chemo-resistance through transcriptionally activating ITGB4. Mol Cell Biochem 2024; 479:665-677. [PMID: 37160555 DOI: 10.1007/s11010-023-04737-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/09/2023] [Indexed: 05/11/2023]
Abstract
Cancer-associated fibroblasts (CAFs) have been proved to facilitate colorectal cancer (CRC) development, either with boosting chemo-resistance by communicating with CRC cells in the tumor microenvironment. However, the underlying molecular mechanisms remain largely unclear. Relative expressions of FOSL1 and ITGB4, either with their correlations in CRC tissues, were assessed using qRT-PCR analysis. Also, Kaplan-Meier survival analysis was employed for evaluating the prognosis. Identification of CAFs was determined by the detection of specific makers (α-SMA, FAP, and FSP1) using western blot and immunofluorescence staining. Cell proliferation, self-renewal capacity, and cell apoptosis were estimated by CCK-8, sphere-formation, and flow cytometry assays. Transcriptional regulation of FOSL1 on integrin β4 (ITGB4) was confirmed using ChIP and dual-luciferase reporter assays. Increased FOSL1 and ITGB4 in CRC tissues were both positively correlated with the poor prognosis of CRC patients. Interestingly, FOSL1 was enriched in the CAFs isolated from CRC stroma, instead of ITGB4. CRC cells under a co-culture system with CAFs-conditioned medium (CAFs-CM) exhibited increased FOSL1, promotive cell proliferation, and reduced apoptosis, while these effects could be blocked by exosome inhibitor (GW4869). Moreover, CAFs-derived exosomal FOSL1 was validated to enhance proliferative ability and oxaliplatin resistance of CRC cells. Our results uncovered that CAFs-derived exosomes could transfer FOSL1 to CRC cells, thereby promoting CRC cell proliferation, stemness, and oxaliplatin resistance by transcriptionally activating ITGB4.
Collapse
Affiliation(s)
- Shanshan Lin
- Department of Rehabilitation Medicine, Jiangmen Central Hospital, Jiangmen, 529099, Guangdong Province, China
| | - Bo Zhu
- Department of Surgical Oncology, Zhongshan City People's Hospital, No. 2 Sunwen East Road, Zhongshan City, Guangdong Province, China.
| |
Collapse
|
48
|
Chen S, Wang C, Meng Y, Li P, Pan Y, He M, Ni X. Nanofabrications of Erythrocyte Membrane-Coated Telmisartan Delivery System Effective for Radiosensitivity of Tumor Cells in Mice Model. Int J Nanomedicine 2024; 19:1487-1508. [PMID: 38380147 PMCID: PMC10878400 DOI: 10.2147/ijn.s441418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024] Open
Abstract
Background Radiation stimulates the secretion of tumor stroma and induces resistance, recurrence, and metastasis of stromal-vascular tumors during radiotherapy. The proliferation and activation of tumor-associated fibroblasts (TAFs) are important reasons for the production of tumor stroma. Telmisartan (Tel) can inhibit the proliferation and activation of TAFs (resting TAFs), which may promote radiosensitization. However, Tel has a poor water solubility. Methods In this study, self-assembled telmisartan nanoparticles (Tel NPs) were prepared by aqueous solvent diffusion method to solve the insoluble problem of Tel and achieve high drug loading of Tel. Then, erythrocyte membrane (ECM) obtained by hypotonic lysis was coated on the surface of Tel NPs (ECM/Tel) for the achievement of in vivo long circulation and tumor targeting. Immunofluorescence staining, western blot and other biological techniques were used to investigate the effect of ECM/Tel on TAFs activation inhibition (resting effect) and mechanisms involved. The multicellular spheroids (MCSs) model and mouse breast cancer cells (4T1) were constructed to investigate the effect of ECM/Tel on reducing stroma secretion, alleviating hypoxia, and the corresponding promoting radiosensitization effect in vitro. A mouse orthotopic 4T1 breast cancer model was constructed to investigate the radiosensitizing effect of ECM/Tel on inhibiting breast cancer growth and lung metastasis of breast cancer. Results ECM/Tel showed good physiological stability and tumor-targeting ability. ECM/Tel could rest TAFs and reduce stroma secretion, alleviate hypoxia, and enhance penetration in tumor microenvironment. In addition, ECM/Tel arrested the cell cycle of 4T1 cells to the radiosensitive G2/M phase. In mouse orthotopic 4T1 breast cancer model, ECM/Tel played a superior role in radiosensitization and significantly inhibited lung metastasis of breast cancer. Conclusion ECM/Tel showed synergistical radiosensitization effect on both the tumor microenvironment and tumor cells, which is a promising radiosensitizer in the radiotherapy of stroma-vascular tumors.
Collapse
Affiliation(s)
- Shaoqing Chen
- Department of Radiotherapy, the Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213003, People’s Republic of China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, 213003, People’s Republic of China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, 213164, People’s Republic of China
| | - Yanyan Meng
- Department of Radiotherapy, the Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213003, People’s Republic of China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, 213003, People’s Republic of China
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, 213164, People’s Republic of China
| | - Pengyin Li
- Department of Radiotherapy, the Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213003, People’s Republic of China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, 213003, People’s Republic of China
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, 213164, People’s Republic of China
| | - Yiwen Pan
- Department of Radiotherapy, the Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213003, People’s Republic of China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, 213003, People’s Republic of China
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, 213164, People’s Republic of China
| | - Mu He
- Department of Radiotherapy, the Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213003, People’s Republic of China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, 213003, People’s Republic of China
| | - Xinye Ni
- Department of Radiotherapy, the Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213003, People’s Republic of China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, 213003, People’s Republic of China
| |
Collapse
|
49
|
Yin SY, Liu YJ, Li JP, Liu J. Overexpression of FERM Domain Containing Kindlin 2 (FERMT2) in Fibroblasts Correlates with EMT and Immunosuppression in Gastric Cancer. Int J Genomics 2024; 2024:4123737. [PMID: 38352691 PMCID: PMC10864055 DOI: 10.1155/2024/4123737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/26/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
The mesenchymal feature, dominated by epithelial mesenchymal transition (EMT) and stromal cell activation, is one of the main reasons for the aggressive nature of tumors, yet it remains poorly understood. In gastric cancer (GC), the fermitin family homolog-2 (FERMT2) is involved in macrophage signaling, promoting migration and invasion. However, the function of FERMT2 in fibroblasts remains unclear. Here, we demonstrated that downregulation of FERMT2 expression can block EMT in GC cells by inhibiting fibroblast activation in vitro. Furthermore, we found that, in addition to the known pathways, fibroblast-derived FERMT2 promotes M2-like macrophage growth and that in human GC samples, there is a strong positive correlation between FERMT2 and CD163 and CD206 levels. Notably, high FERMT2 expression was significantly associated with poor clinical outcomes and was upregulated in patients with advanced disease. Taken together, our results provide evidence that the fibroblast-FERMT2-EMT-M2 macrophage axis plays a critical role in the GC mesenchymal phenotype and may be a promising target for the treatment of advanced GC.
Collapse
Affiliation(s)
- Sheng-yan Yin
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Second Chinese Medicine Hospital, Nanjing, Jiangsu 210029, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Yuan-jie Liu
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Second Chinese Medicine Hospital, Nanjing, Jiangsu 210029, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Jie-pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jian Liu
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Second Chinese Medicine Hospital, Nanjing, Jiangsu 210029, China
| |
Collapse
|
50
|
Franchi M, Piperigkou Z, Mastronikolis NS, Karamanos N. Extracellular matrix biomechanical roles and adaptation in health and disease. FEBS J 2024; 291:430-440. [PMID: 37612040 DOI: 10.1111/febs.16938] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2023] [Accepted: 08/22/2023] [Indexed: 08/25/2023]
Abstract
Extracellular matrices (ECMs) are dynamic 3D macromolecular networks that exhibit structural characteristics and composition specific to different tissues, serving various biomechanical and regulatory functions. The interactions between ECM macromolecules such as collagen, elastin, glycosaminoglycans (GAGs), proteoglycans (PGs), fibronectin, and laminin, along with matrix effectors and water, contribute to the unique cellular and tissue functional properties during organ development, tissue homoeostasis, remodeling, disease development, and progression. Cells adapt to environmental changes by adjusting the composition and array of ECM components. ECMs, forming the 3D bioscaffolds of our body, provide mechanical support for tissues and organs and respond to the environmental variables influencing growth and final adult body shape in mammals. Different cell types display distinct adaptations to the respective ECM environments. ECMs regulate biological processes by controlling the diffusion of infections and inflammations, sensing and adapting to external stimuli and gravity from the surrounding habitat, and, in the context of cancer, interplaying with and regulating cancer cell invasion and drug resistance. Alterations in the ECM composition in pathological conditions drive adaptive responses of cells and could therefore result in abnormal cell behavior and tissue dysfunction. Understanding the biomechanical functionality, adaptation, and roles of distinct ECMs is essential for research on various pathologies, including cancer progression and multidrug resistance, which is of crucial importance for developing targeted therapies. In this Viewpoint article, we critically present and discuss specific biomechanical functions of ECMs and regulatory adaptation mechanisms in both health and disease, with a particular focus on cancer progression.
Collapse
Affiliation(s)
- Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Zoi Piperigkou
- Department of Chemistry, Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Nicholas S Mastronikolis
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, University of Patras, Greece
| | - Nikos Karamanos
- Department of Chemistry, Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|