1
|
Miao C, Sun R, Ji D, Wu M, Fu Q, Mei L, Wu Z. Mechanism of the GALNT family proteins in regulating tumorigenesis and development of lung cancer (Review). Mol Clin Oncol 2025; 22:37. [PMID: 40083861 PMCID: PMC11904754 DOI: 10.3892/mco.2025.2832] [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: 09/11/2024] [Accepted: 02/20/2025] [Indexed: 03/16/2025] Open
Abstract
Lung cancer is one of the most common and lethal malignant tumors. Currently, surgical resection is the most effective treatment for early-stage lung cancer, and often results in favorable recovery outcomes. Therefore, early detection and control of lung cancer occurrence, invasion and metastasis are crucial for improving patient survival rates. Identifying tumor markers for lung cancer plays a vital role in facilitating early detection and control of its progression. The GALNT family proteins are enzymes that regulate the initial step in mucin O-glycan synthesis. It has been revealed that the expression of polypeptide N-acetyl-galactosamine-transferase (GALNT) family members is dysregulated in various tumors, and is closely associated with tumorigenesis, tumor cell growth, metastasis, adhesion, and serves as an important early indicator of tumor development. The present review compiles and analyzes findings concerning the role of GALNT family proteins in regulating lung cancer, with the goals of elucidating their mechanisms in lung cancer occurrence and progression and providing insights for improving the prognosis and therapeutic treatment of patients with lung cancer.
Collapse
Affiliation(s)
- Changchun Miao
- Department of Cardiothoracic Surgery, Mingguang People's Hospital, Mingguang, Anhui 239400, P.R. China
| | - Ronggui Sun
- Department of Cardiothoracic Surgery, Mingguang People's Hospital, Mingguang, Anhui 239400, P.R. China
| | - Deyu Ji
- Department of Cardiothoracic Surgery, Mingguang People's Hospital, Mingguang, Anhui 239400, P.R. China
| | - Min Wu
- Department of Cardiothoracic Surgery, Mingguang People's Hospital, Mingguang, Anhui 239400, P.R. China
| | - Qigui Fu
- Department of Cardiothoracic Surgery, Mingguang People's Hospital, Mingguang, Anhui 239400, P.R. China
| | - Liangliang Mei
- Department of Cardiothoracic Surgery, Mingguang People's Hospital, Mingguang, Anhui 239400, P.R. China
| | - Zhiyong Wu
- Department of Cardiothoracic Surgery, Mingguang People's Hospital, Mingguang, Anhui 239400, P.R. China
| |
Collapse
|
2
|
Guo R, Xie X, Ren Q, Liew PX. New insights on extramedullary granulopoiesis and neutrophil heterogeneity in the spleen and its importance in disease. J Leukoc Biol 2025; 117:qiae220. [PMID: 39514106 DOI: 10.1093/jleuko/qiae220] [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: 09/03/2024] [Indexed: 11/16/2024] Open
Abstract
Neutrophils are traditionally viewed as uncomplicated exterminators that arrive quickly at sites of infection, kill pathogens, and then expire. However, recent studies employing modern transcriptomics coupled with novel imaging modalities have discovered that neutrophils exhibit significant heterogeneity within organs and have complex functional roles ranging from tissue homeostasis to cancer and chronic pathologies. This has revised the view that neutrophils are simplistic butchers, and there has been a resurgent interest in neutrophils. The spleen was described as a granulopoietic organ more than 4 decades ago, and studies indicate that neutrophils are briefly retained in the spleen before returning to circulation after proliferation. Transcriptomic studies have discovered that splenic neutrophils are heterogeneous and distinct compared with those in blood. This suggests that a unique hematopoietic niche exists in the splenic microenvironment, i.e., capable of programming neutrophils in the spleen. During severe systemic inflammation with an increased need of neutrophils, the spleen can adapt by producing neutrophils through emergency granulopoiesis. In this review, we describe the structure and microanatomy of the spleen and examine how cells within the splenic microenvironment help to regulate splenic granulopoiesis. A focus is placed on exploring the increase in splenic granulopoiesis to meet host needs during infection and inflammation. Emerging technologies such as single-cell RNA sequencing, which provide valuable insight into splenic neutrophil development and heterogeneity, are also discussed. Finally, we examine how tumors subvert this natural pathway in the spleen to generate granulocytic suppressor cells to promote tumor growth.
Collapse
Affiliation(s)
- Rongxia Guo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan, Hubei 430071, China
| | - Xuemei Xie
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 77 Ave Louis Pasteur, Boston, MA 02115, United States
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Heping District, Tianjin 300020, China
- Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences, 288 Nanjing Road, Heping District, Tianjin 300020, China
| | - Pei Xiong Liew
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, United States
- Department of Cellular Biology and Anatomy, Augusta University, 1434 Laney Walker Blvd, Augusta, GA 30912, United States
| |
Collapse
|
3
|
Liu X, Tao P, Su H, Li Y. Machine learning-random forest model was used to construct gene signature associated with cuproptosis to predict the prognosis of gastric cancer. Sci Rep 2025; 15:4170. [PMID: 39905263 PMCID: PMC11794614 DOI: 10.1038/s41598-025-88812-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Gastric cancer (GC) is one of the most common tumors; one of the reasons for its poor prognosis is that GC cells can resist normal cell death process and therefore develop distant metastasis. Cuproptosis is a novel type of cell death and a limited number of studies have been conducted on the relationship between cuproptosis-related genes (CRGs) in GC. The purpose of the present study was to establish a prognostic model of CRGs and provide directions for the diagnosis and treatment of GC. Transcriptome and clinical data of patients with GC were collected from The Cancer Genome Atlas and Gene Expression Omnibus datasets. Single sample gene set enrichment analysis (GSEA) and the randomized forest method were used to establish the prognostic model. Kaplan-Meier survival curve, receiver operating characteristics diagram and a nomogram were used to evaluate the reliability of the model. GSEA and gene set variation analysis (GSVA) were used to examine enrichment pathways between high and low risk groups. Finally, immunohistochemical analysis was used to examine ephrin 4 (EFNA4) expression in GC samples and determine the prognosis of patients with GC based on the expression pattern of EFNA4. A group of 7 predictive models (RTKN2, INO80B, EFNA4, ELF2, MUSTN, KRTAP4, and ARHGEF40) was established which were correlated with CRGs. This model can be used as an independent prognostic factor to predict the prognosis of patients with GC. GSEA and GSVA results indicated that high risk patients with GC were mainly associated with the enrichment of ANGIOGENESIS and TGF_BETA_SIGNALING pathways. Finally, EFNA4 expression in GC was significantly higher than that in normal tissues, and patients with GC and high EFNA4 expression exhibited improved prognosis. In conclusion, the prognosis model based on CRGs could be used as the basis for predicting the potential prognosis of patients with GC and provide new insights for the treatment of GC.
Collapse
Affiliation(s)
- Xiaolong Liu
- The First School of Clinical Medical, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, Gansu, People's Republic of China
- Department of Science and Education, The Third People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China
| | - Pengxian Tao
- Cadre Ward of General Surgery Department, Gansu Provincial Hospital, 204 Donggang West Road, Chengguan, Lanzhou, 730000, Gansu, People's Republic of China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Gansu, 730000, People's Republic of China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, People's Republic of China
| | - He Su
- Cadre Ward of General Surgery Department, Gansu Provincial Hospital, 204 Donggang West Road, Chengguan, Lanzhou, 730000, Gansu, People's Republic of China.
| | - Yulan Li
- The First School of Clinical Medical, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
4
|
Neumeyer V, Chavan P, Steiger K, Ebert O, Altomonte J. Cross-Talk Between Tumor Cells and Stellate Cells Promotes Oncolytic VSV Activity in Intrahepatic Cholangiocarcinoma. Cancers (Basel) 2025; 17:514. [PMID: 39941881 PMCID: PMC11816849 DOI: 10.3390/cancers17030514] [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: 12/19/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
As the mechanisms underlying tumorigenesis become better understood, the dynamic roles of cellular components of the tumor microenvironment, and their cross-talk with tumor cells, have come to light as key drivers of disease progression and have emerged as important targets of new cancer therapies. In the field of oncolytic virus (OV) therapy, stromal cells have been considered as potential barriers to viral spread, thus limiting virus replication and therapeutic outcome. However, new evidence indicates that intratumoral fibroblasts could support virus replication. We have demonstrated in a rat model of stromal-rich intrahepatic cholangiocarcinoma (CCA) that vesicular stomatitis virus (VSV) can be localized within intratumoral hepatic stellate cells (HSCs), in addition to tumor cells, when the virus was applied via hepatic arterial infusion. Furthermore, VSV was shown to efficiently kill CCA cells and activated HSCs, and co-culture of CCA and HSCs increased viral titers. Interestingly, this effect is also observed when each cell type is cultured alone in a conditioned medium of the other cell type, indicating that secreted cell factors are at least partially responsible for this phenomenon. Partial reduction in sensitivity to type I interferons was observed in co-culture systems, providing a possible mechanism for the increased viral titers. Together, the results indicate that targeting activated HSCs with VSV could provide an additional mechanism of OV therapy, which, until now has not been considered. Furthermore, these findings suggest that VSV is a potentially powerful therapeutic agent for stromal-rich tumors, such as CCA and pancreatic cancer, both of which are very difficult to treat with conventional therapy and have a very poor prognosis.
Collapse
Affiliation(s)
- Victoria Neumeyer
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| | - Purva Chavan
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- Department of Pathology, Technical University of Munich, 81675 Munich, Germany
| | - Oliver Ebert
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| | - Jennifer Altomonte
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
5
|
Famta P, Shah S, Vambhurkar G, Pandey G, Bagasariya D, Kumar KC, Prasad SB, Shinde A, Wagh S, Srinivasarao DA, Kumar R, Khatri DK, Asthana A, Srivastava S. Amelioration of breast cancer therapies through normalization of tumor vessels and microenvironment: paradigm shift to improve drug perfusion and nanocarrier permeation. Drug Deliv Transl Res 2025; 15:389-406. [PMID: 39009931 DOI: 10.1007/s13346-024-01669-9] [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] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer among women. Chemo-, immune- and photothermal therapies are employed to manage BC. However, the tumor microenvironment (TME) prevents free drugs and nanocarriers (NCs) from entering the tumor premises. Formulation scientists rely on enhanced permeation and retention (EPR) to extravasate NCs in the TME. However, recent research has demonstrated the inconsistent nature of EPR among different patients and tumor types. In addition, angiogenesis, high intra-tumor fluid pressure, desmoplasia, and high cell and extracellular matrix density resist the accumulation of NCs in the TME. In this review, we discuss TME normalization as an approach to improve the penetration of drugs and NCSs in the tumor premises. Strategies such as normalization of tumor vessels, reversal of hypoxia, alleviation of high intra-tumor pressure, and infiltration of lymphocytes for the reversal of therapy failure have been discussed in this manuscript. Strategies to promote the infiltration of anticancer immune cells in the TME after vascular normalization have been discussed. Studies strategizing time points to administer TME-normalizing agents are highlighted. Mechanistic pathways controlling the angiogenesis and normalization processes are discussed along with the studies. This review will provide greater tumor-targeting insights to the formulation scientists.
Collapse
Affiliation(s)
- Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Deepkumar Bagasariya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Kondasingh Charan Kumar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Sajja Bhanu Prasad
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Akshay Shinde
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Suraj Wagh
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
- Molecular and Cellular Biology Laboratory, Department of Pharmacology, Nims Institute of Pharmacy, Nims University, Jaipur, Rajasthan, India
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
6
|
Gu JH, Chae MY, Choi JJ, Duc TC, Son CG, Lee EJ. Deer antler velvet (Cervus elaphus sibiricus) promotes fracture healing via partial BMP2-Smad mediated osteoblast differentiation. J Orthop Surg Res 2025; 20:70. [PMID: 39833827 PMCID: PMC11744865 DOI: 10.1186/s13018-024-05426-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND CERVUS ELAPHUS SIBIRICUS: (CES) has been traditionally used in Korean clinics to promote fracture healing based on its function of tonifying the kidneys and strengthening bones. However, experimental data supporting its efficacy are still insufficient. The aim of this study investigated the bone-union properties of CES in a femoral fracture animal model and its corresponding molecular mechanisms. METHODS Fifty-four C57BL/6 male mice underwent femoral shaft fracture by Bonnarens and Einhorn's method, subsequently receiving a water extract of CES (200 mg/kg/day, daily) for 7 and 14 days. Safranin O staining and immunohistochemistry of the fracture region were conducted against transforming growth factor-β (TGF-β), bone morphogenetic protein 2 (BMP2), and osterix. MG63 cells used to examine the underlying mechanisms of CES focused on BMP2-Smad pathway-related osteogenesis. RESULTS CES administration led to earlier union of the fractured bones, supported by Safranin O staining of the fracture region, demonstrating significantly increased cartilage formation day on 7 and a rapidly decreased cartilage area due to callus formation day on 14. CES administration also significantly upregulated the expression of TGF-β1 day 7, BMP 2, and osterix day 14 at the fracture site and also up-regulated alkaline phosphatase (ALP) activity, calcium deposition, and the phosphorylation of Smad in MG63 cells. CONCLUSIONS CES promotes fracture healing by promoting osteoblastogenesis via a partial BMP2-Smad pathway.
Collapse
Affiliation(s)
- Ji Hyang Gu
- Department of Korean Rehabilitation Medicine, Cheonan Korean Medicine Hospital of Daejeon University, Cheonan, 31099, South Korea
| | - Min-Young Chae
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Daejeon University, Daejeon, 35235, South Korea
| | - Jeong June Choi
- Laboratory of Molecular Medicine, College of Korean Medicine, Daejeon University, Daejeon, 34520, South Korea
| | - Thanh Chu Duc
- College of Pharmacy, Chungnam National University, Daejeon, 34134, South Korea
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Ha Noi, Viet Nam
| | - Chang Gue Son
- Institute of Bioscience & Integrative Medicine, Dunsan Hospital of Daejeon University, Daejeon, South Korea
| | - Eun-Jung Lee
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Daejeon University, Daejeon, 35235, South Korea.
| |
Collapse
|
7
|
Qin G, Shao X, Liu X, Xu J, Wang X, Wang W, Gao L, Liang Y, Xie L, Su D, Yang H, Zhou W, Fang X. A signaling molecule from intratumor bacteria promotes trastuzumab resistance in breast cancer cells. Proc Natl Acad Sci U S A 2025; 122:e2421710122. [PMID: 39786928 PMCID: PMC11745319 DOI: 10.1073/pnas.2421710122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025] Open
Abstract
Emerging evidence indicates that intratumor bacteria exist as an active and specific tumor component in many tumor types beyond digestive and respiratory tumors. However, the biological impact and responsible molecules of such local bacteria-tumor direct interaction on cancer therapeutic response remain poorly understood. Trastuzumab is among the most commonly used drugs targeting the receptor tyrosine-protein kinase erbB-2 (ErbB2) in breast cancer, but its resistance is inevitable, severely limiting its clinical effectiveness. Here, we demonstrate that the quorum-sensing signaling molecule N-(3-oxo-dodecanoyl) homoserine lactone (3oc), a chemical compound released by Pseudomonas aeruginosa (P. aeruginosa), one tumor-resident bacteria with a relative high abundance in breast cancer, promotes breast cancer cell resistance to trastuzumab. Mechanically, 3oc directly leads to spontaneous dimerization of the transforming growth factor β (TGF-β) type II serine/threonine kinase receptor on the cell membrane in a ligand-independent manner. The 3oc-induced TGF-β signaling subsequently triggers ErbB2 phosphorylation and its downstream target activation, overcoming the inhibition effect of trastuzumab on ErbB2. With specific real-time qPCR, fluorescence in situ hybridization imaging, and liquid chromatography ionization tandem mass spectrometry analyses of clinical samples, we confirmed that P. aeruginosa and its signaling molecule 3oc exist in breast cancer tissues and there is a clinical correlation between P. aeruginosa colonization and trastuzumab resistance. This work expands the biological functions of intratumor bacteria in cancer treatment responsiveness and provides a unique perspective for overcoming trastuzumab resistance.
Collapse
Affiliation(s)
- Gege Qin
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua-Peking Center for Life Sciences, Institute of Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing100084, China
| | - Xiying Shao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Xiaolong Liu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Jiachao Xu
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Wenxi Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
| | - Lu Gao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Yuxin Liang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Lina Xie
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Dan Su
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Hongwei Yang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
| | - Wei Zhou
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
8
|
Liu J, Zhang B, Huang B, Zhang K, Guo F, Wang Z, Shang D. A stumbling block in pancreatic cancer treatment: drug resistance signaling networks. Front Cell Dev Biol 2025; 12:1462808. [PMID: 39872846 PMCID: PMC11770040 DOI: 10.3389/fcell.2024.1462808] [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: 07/10/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
The primary node molecules in the cell signaling network in cancer tissues are maladjusted and mutated in comparison to normal tissues, which promotes the occurrence and progression of cancer. Pancreatic cancer (PC) is a highly fatal cancer with increasing incidence and low five-year survival rates. Currently, there are several therapies that target cell signaling networks in PC. However, PC is a "cold tumor" with a unique immunosuppressive tumor microenvironment (poor effector T cell infiltration, low antigen specificity), and targeting a single gene or pathway is basically ineffective in clinical practice. Targeted matrix therapy, targeted metabolic therapy, targeted mutant gene therapy, immunosuppressive therapy, cancer vaccines, and other emerging therapies have shown great therapeutic potential, but results have been disappointing. Therefore, we summarize the identified and potential drug-resistant cell signaling networks aimed at overcoming barriers to existing PC therapies.
Collapse
Affiliation(s)
- Jinming Liu
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Biao Zhang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingqian Huang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Westlake University, Hangzhou, China
| | - Kexin Zhang
- Central Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fujia Guo
- Central Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhizhou Wang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Pancreas and Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Xu Y, Lv J, Liu Y, Du J, Luo C, Wang Y, Liu L, Sakurai K, Tang Z, Chen X. Coagulation-Targeted TGF-β Signaling Pathway Inhibitor Nanomedicine for Inhibiting the Growth and Lung Metastasis of Breast Cancer. NANO LETTERS 2025; 25:504-513. [PMID: 39680715 DOI: 10.1021/acs.nanolett.4c05355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The transforming growth factor β (TGF-β) signaling pathway exerts a dual role in oncogenesis, acting as a suppressor in healthy and early stage neoplastic tissues while promoting malignancy and metastasis in advanced cancers. Tumor hemorrhage further exacerbates TGF-β-mediated metastasis by up-regulating its expression. Here, a coagulation-targeting peptide (A15)-decorated TGF-β inhibitor nanomedicine (A15-LY-NPs) was developed. The tumor colonization assays showed that the nanomedicine reduced 4T1-luc cell colonization in normal tissues. When combined with a vascular disrupting agent, A15-LY-NPs demonstrated three times greater drug accumulation in the tumor at 24 h compared to the control and showed a 93.7% tumor suppression rate in 4T1 tumors initiated at ∼500 mm3, significantly attenuating metastatic spread to the lungs and liver. This study presents an innovative approach for the precise and efficient delivery of TGF-β inhibitors to tumors, offering the potential to augment the efficacy of cancer therapeutics.
Collapse
Affiliation(s)
- Yajun Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jianlin Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Ya Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Jincheng Du
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Chuwen Luo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Ying Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Linlin Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1 Hibikino, Kitakyushu 808-0135, Japan
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
10
|
Maldonado H, Dreger M, Bedgood LD, Kyriakou T, Wolanska KI, Rigby ME, Marotta VE, Webster JM, Wang J, Rusilowicz-Jones EV, Marshall JF, Coulson JM, Macpherson IR, Hurlstone A, Morgan MR. A trafficking regulatory subnetwork governs α Vβ 6 integrin-HER2 cross-talk to control breast cancer invasion and drug resistance. SCIENCE ADVANCES 2024; 10:eadk9944. [PMID: 39630893 PMCID: PMC11616693 DOI: 10.1126/sciadv.adk9944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/31/2024] [Indexed: 12/07/2024]
Abstract
HER2 and αVβ6 integrin are independent predictors of breast cancer survival and metastasis. We identify an αVβ6/HER2 cross-talk mechanism driving invasion, which is dysregulated in drug-resistant HER2+ breast cancer cells. Proteomic analyses reveal ligand-bound αVβ6 recruits HER2 and a trafficking subnetwork, comprising guanosine triphosphatases RAB5 and RAB7A and the Rab regulator guanine nucleotide dissociation inhibitor 2 (GDI2). The RAB5/RAB7A/GDI2 functional module mediates direct cross-talk between αVβ6 and HER2, affecting receptor trafficking and signaling. Acute exposure to trastuzumab increases recruitment of the subnetwork to αVβ6, but trastuzumab resistance decouples GDI2 recruitment. GDI2, RAB5, and RAB7A cooperate to regulate migration and transforming growth factor-β activation to promote invasion. However, these mechanisms are dysregulated in trastuzumab-resistant cells. In patients, RAB5A, RAB7A, and GDI2 expression correlates with patient survival and αVβ6 expression predicts relapse following trastuzumab treatment. Thus, the RAB5/RAB7A/GDI2 subnetwork regulates αVβ6-HER2 cross-talk to drive breast cancer invasion but is subverted in trastuzumab-resistant cells to drive αVβ6-independent and HER2-independent tumor progression.
Collapse
Affiliation(s)
- Horacio Maldonado
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Marcel Dreger
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Lara D. Bedgood
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Theano Kyriakou
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Katarzyna I. Wolanska
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Megan E. Rigby
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Valeria E. Marotta
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Justine M. Webster
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Jun Wang
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Emma V. Rusilowicz-Jones
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Judy M. Coulson
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Iain R. Macpherson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| | - Adam Hurlstone
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Mark R. Morgan
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| |
Collapse
|
11
|
Carvalho MI, Silva-Carvalho R, Prada J, Pinto C, Gregório H, Lobo L, Pires I, Queiroga FL. TGFβ in malignant canine mammary tumors: relation with angiogenesis, immunologic markers and prognostic role. Vet Q 2024; 44:1-12. [PMID: 39165025 PMCID: PMC11340227 DOI: 10.1080/01652176.2024.2390941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/22/2024] Open
Abstract
Transforming growth factor-β (TGFβ) and FoxP3 regulatory T cells (Treg) are involved in human breast carcinogenesis. This topic is not well documented in canine mammary tumors (CMT). In this work, the tumoral TGFβ expression was assessed by immunohistochemistry in 67 malignant CMT and its correlation to previously determined FoxP3, VEGF, and CD31 markers and other clinicopathologic parameters was evaluated. The high levels of TGFβ were statistically significantly associated with skin ulceration, tumor necrosis, high histological grade of malignancy (HGM), presence of neoplastic intravascular emboli and presence of lymph node metastases. The observed levels of TGFβ were positively correlated with intratumoral FoxP3 (strong correlation), VEGF (weak correlation) and CD31 (moderate correlation). Tumors that presented a concurrent high expression of TGFβ/FoxP3, TGFβ/VEGF, and TGFβ/CD31 markers were statistically significantly associated with parameters of tumor malignancy (high HGM, presence of vascular emboli and nodal metastasis). Additionally, shorter overall survival (OS) time was statistically significantly associated with tumors with an abundant TGFβ expression and with concurrent high expression of TGFβ/FoxP3, TGFβ/VEGF, and TGFβ/CD31. The presence of lymph node metastasis increased 11 times the risk of disease-related death, arising as an independent predictor of poor prognosis in the multivariable analysis. In conclusion, TGFβ and Treg cells seem involved in tumor progression emerging as potential therapeutic targets for future immunotherapy studies.
Collapse
Affiliation(s)
- Maria Isabel Carvalho
- MVET Research in Veterinary Medicine. Faculty of Veterinary Medicine, Lusófona University – Lisbon Centre, Lisboa, Portugal
| | - Ricardo Silva-Carvalho
- CEB – Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
| | - Justina Prada
- Veterinary and Animal Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Carla Pinto
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Hugo Gregório
- Anicura Centro Hospitalar Veterinário, Porto, Portugal
| | - Luis Lobo
- MVET Research in Veterinary Medicine. Faculty of Veterinary Medicine, Lusófona University – Lisbon Centre, Lisboa, Portugal
- Onevet Hospital Veterinário do Porto, Porto, Portugal
- Center for the Study of Animal Sciences, CECA-ICETA, University of Porto, Portugal
| | - Isabel Pires
- Veterinary and Animal Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Felisbina L. Queiroga
- Veterinary and Animal Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Center for the Study of Animal Sciences, CECA-ICETA, University of Porto, Portugal
| |
Collapse
|
12
|
Zheng Y, Li L, Cai W, Li L, Zhang R, Huang W, Cao Y. Unveiling the role of TGF-β signaling pathway in breast cancer prognosis and immunotherapy. Front Oncol 2024; 14:1488137. [PMID: 39664194 PMCID: PMC11631921 DOI: 10.3389/fonc.2024.1488137] [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: 08/29/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction The TGF-β signaling pathway (TSP) is pivotal in tumor progression. Nonetheless, the connection between genes associated with the TSP and the clinical outcomes of breast cancer, as well as their impact on the tumor microenvironment and immunotherapeutic responses, remains elusive. Methods Breast cancer transcriptomic and single-cell sequencing data were obtained from the The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. We identified 54 genes associated with the TSP from the Molecular Signatures Database (MSigDB) and analyzed both data types to evaluate TSP activity. Using weighted gene co-expression network analysis (WGCNA), we identified modules linked to TSP activity. To assess patient risk, we used 101 machine learning algorithms to develop an optimal TGF-β pathway-related prognostic signature (TSPRS). We then examined immune activity and response to immune checkpoint inhibitors and chemotherapy in these groups. Finally, we validated ZMAT3 expression levels clinically and confirmed its relevance in breast cancer using CCK-8 and migration assays. Results At the single-cell level, TSP activity was most notable in endothelial cells, with higher activity in normal tissues compared to tumors. TSPRS was developed. This signature's accuracy was confirmed through internal and external validations. A nomogram incorporating the TSPRS was created to improve prediction accuracy. Further studies showed that breast cancer patients categorized as low-risk by the TSPRS had higher immune phenotype scores and more immune cell infiltration, leading to better prognosis and enhanced immunotherapy response. Additionally, a strong link was found between the TSPRS risk score and the effectiveness of anti-tumor agents. Silencing the ZMAT3 gene in the TSPRS significantly reduced the proliferation and invasiveness of breast cancer cells. Discussion Our study developed a TSPRS, which emerges as a potent predictive instrument for the prognosis of breast cancer, offering novel perspectives on the immunotherapeutic approach to the disease.
Collapse
Affiliation(s)
- Yifan Zheng
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenqian Cai
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lin Li
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenbin Huang
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yulun Cao
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Letson J, Ren G, Zheng X, Sweef O, Corcino YL, Furuta S. Reduced S-nitrosylation of TGFβ1 elevates its binding affinity toward the receptor and promotes fibrogenic signaling in the breast. J Biol Chem 2024; 300:108011. [PMID: 39571651 PMCID: PMC11699740 DOI: 10.1016/j.jbc.2024.108011] [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: 01/12/2024] [Revised: 08/19/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Transforming Growth Factor β (TGFβ) is a pleiotropic cytokine closely linked to tumors. Previously, we pharmacologically inhibited basal nitric oxide (NO) production in healthy mammary glands and found that this induced precancerous progression accompanied by upregulation of TGFβ and desmoplasia. In the present study, we tested whether NO directly S-nitrosylates (forms an NO-adduct at a cysteine residue) TGFβ for inhibition, whereas reduction of NO denitrosylates TGFβ for de-repression. We introduced mutations to 3 C-terminal cysteines of TGFβ1 which were predicted to be S-nitrosylated. We found that these mutations indeed impaired S-nitrosylation of TGFβ1 and shifted the binding affinity towards the receptor from the latent complex. Furthermore, in silico structural analyses predicted that these S-nitrosylation-defective mutations strengthen the dimerization of mature protein, whereas S-nitrosylation-mimetic mutations weaken the dimerization. Such differences in dimerization dynamics of TGFβ1 by denitrosylation/S-nitrosylation likely account for the shift of the binding affinities toward the receptor versus latent complex. Our findings, for the first time, unravel a novel mode of TGFβ regulation based on S-nitrosylation or denitrosylation of the protein.
Collapse
Affiliation(s)
- Joshua Letson
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, Ohio, USA; Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, Ohio, USA
| | - Gang Ren
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, Ohio, USA; Department of Surgery, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, Ohio, USA
| | - Xunzhen Zheng
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, Ohio, USA
| | - Osama Sweef
- Department of Medicine, MetroHealth Medical Center, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Faculty of Science, Department of Zoology, Tanta University, Tanta, Egypt
| | - Yalitza Lopes Corcino
- Department of Medicine, MetroHealth Medical Center, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Saori Furuta
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, Ohio, USA; Department of Medicine, MetroHealth Medical Center, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.
| |
Collapse
|
14
|
Aoki K, Higuchi T, Akieda Y, Matsubara K, Ohkawa Y, Ishitani T. Mechano-gradients drive morphogen-noise correction to ensure robust patterning. SCIENCE ADVANCES 2024; 10:eadp2357. [PMID: 39546611 PMCID: PMC11567007 DOI: 10.1126/sciadv.adp2357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Morphogen gradients instruct cells to pattern tissues. Although the mechanisms by which morphogens transduce chemical signals have been extensively studied, the roles and regulation of the physical communication between morphogen-receiver cells remain unclear. Here, we show that the Wnt/β-catenin-morphogen gradient, which patterns the embryonic anterior-posterior (AP) axis, generates intercellular tension gradients along the AP axis by controlling membrane cadherin levels in zebrafish embryos. This "mechano-gradient" is used for the cell competition-driven correction of noisy morphogen gradients. Naturally and artificially generated unfit cells, producing noisy Wnt/β-catenin gradients, induce local deformation of the mechano-gradients that activate mechanosensitive calcium channels in the neighboring fit cells, which then secrete annexin A1a to kill unfit cells. Thus, chemo-mechanical interconversion-mediated competitive communication between the morphogen-receiver cells ensures precise tissue patterning.
Collapse
Affiliation(s)
- Kana Aoki
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Taiki Higuchi
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuki Akieda
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kotone Matsubara
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Fukuoka, Fukuoka 812-0054, Japan
| | - Tohru Ishitani
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
15
|
Wang Z, Kometani M, Zeitlin L, Wilnai Y, Kinoshita A, Yoshiura KI, Ninomiya H, Imamura T, Guo L, Xue J, Yan L, Ohashi H, Pretemer Y, Kawai S, Shiina M, Ogata K, Cohn DH, Matsumoto N, Nishimura G, Toguchida J, Miyake N, Ikegawa S. Heterozygous mutations in the straitjacket region of the latency-associated peptide domain of TGFB2 cause Camurati-Engelmann disease type II. J Hum Genet 2024; 69:599-605. [PMID: 39014191 DOI: 10.1038/s10038-024-01274-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024]
Abstract
Camurati-Engelmann disease (CED) is an autosomal dominant bone dysplasia characterized by progressive hyperostosis of the skull base and diaphyses of the long bones. CED is further divided into two subtypes, CED1 and CED2, according to the presence or absence of TGFB1 mutations, respectively. In this study, we used exome sequencing to investigate the genetic cause of CED2 in three pedigrees and identified two de novo heterozygous mutations in TGFB2 among the three patients. Both mutations were located in the region of the gene encoding the straitjacket subdomain of the latency-associated peptide (LAP) of pro-TGF-β2. Structural simulations of the mutant LAPs suggested that the mutations could cause significant conformational changes and lead to a reduction in TGF-β2 inactivation. An activity assay confirmed a significant increase in TGF-β2/SMAD signaling. In vitro osteogenic differentiation experiment using iPS cells from one of the CED2 patients showed significantly enhanced ossification, suggesting that the pathogenic mechanism of CED2 is increased activation of TGF-β2 by loss-of-function of the LAP. These results, in combination with the difference in hyperostosis patterns between CED1 and CED2, suggest distinct functions between TGFB1 and TGFB2 in human skeletal development and homeostasis.
Collapse
Affiliation(s)
- Zheng Wang
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Mitsuhiro Kometani
- Division of Endocrinology and Hypertension, Department of Cardiovascular and Internal Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Leonid Zeitlin
- Pediatric Bone Clinic, Orthopaedic Department, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Yael Wilnai
- Genetic Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Akira Kinoshita
- Department of Human Genetics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Koh-Ichiro Yoshiura
- Department of Human Genetics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hiroko Ninomiya
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Japan
| | - Long Guo
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Jingyi Xue
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Li Yan
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Hirofumi Ohashi
- Division of Medical Genetics, Saitama Children's Medical Genetics, Saitama, Japan
| | - Yann Pretemer
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shunsuke Kawai
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masaaki Shiina
- Department of Biochemistry, Yokohama City University, Yokohama, Japan
| | - Kazuhiro Ogata
- Department of Biochemistry, Yokohama City University, Yokohama, Japan
| | - Daniel H Cohn
- Department of Molecular, Cell, and Developmental Biology, Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Gen Nishimura
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Junya Toguchida
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University, Yokohama, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan.
| |
Collapse
|
16
|
Jang JJ, Kang D, Lee YS, Lee YS. The Versatile Roles of nc886, a Fascinating and Peculiar Regulatory Non-Coding RNA, in Cancer. Int J Mol Sci 2024; 25:10825. [PMID: 39409154 PMCID: PMC11476670 DOI: 10.3390/ijms251910825] [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: 09/04/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
This review concerns nc886, a 101-nucleotide non-coding RNA (ncRNA). Because nc886 is transcribed by RNA polymerase III (Pol III) and contains a CpG island in its promoter region, its expression is regulated by several transcription factors and the DNA methylation status. These features drive nc886 expression in two opposing directions during tumorigenesis. The known function of nc886 is to bind to and modulate the activity of target proteins such as PKR, Dicer, and OAS1. By being differentially expressed during tumorigenesis and interacting with these proteins, nc886 plays a role in tumor surveillance, promotes or suppresses tumorigenesis, and influences the efficacy of cancer therapy. The multiple roles of nc886 have been well-documented in the literature. In this review, we have summarized this literature and critically discussed the roles and mechanisms of action of nc886 in various cancers.
Collapse
Affiliation(s)
- Jiyoung Joan Jang
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea;
- Fluorescence Core Imaging Center, Department of Life Science, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Dongmin Kang
- Fluorescence Core Imaging Center, Department of Life Science, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Yeon-Su Lee
- Division of Rare Cancer, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Yong Sun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea;
| |
Collapse
|
17
|
Lu Y, Zheng J, Lin P, Lin Y, Zheng Y, Mai Z, Chen X, Xia T, Zhao X, Cui L. Tumor Microenvironment-Derived Exosomes: A Double-Edged Sword for Advanced T Cell-Based Immunotherapy. ACS NANO 2024; 18:27230-27260. [PMID: 39319751 DOI: 10.1021/acsnano.4c09190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer progression and immune evasion, partially mediated by the activity of the TME-derived exosomes. These extracellular vesicles are pivotal in shaping immune responses through the transfer of proteins, lipids, and nucleic acids between cells, facilitating a complex interplay that promotes tumor growth and metastasis. This review delves into the dual roles of exosomes in the TME, highlighting both their immunosuppressive functions and their emerging therapeutic potential. Exosomes can inhibit T cell function and promote tumor immune escape by carrying immune-modulatory molecules, such as PD-L1, yet they also hold promise for cancer therapy as vehicles for delivering tumor antigens and costimulatory signals. Additionally, the review discusses the intricate crosstalk mediated by exosomes among various cell types within the TME, influencing both cancer progression and responses to immunotherapies. Moreover, this highlights current challenges and future directions. Collectively, elucidating the detailed mechanisms by which TME-derived exosomes mediate T cell function offers a promising avenue for revolutionizing cancer treatment. Understanding these interactions allows for the development of targeted therapies that manipulate exosomal pathways to enhance the immune system's response to tumors.
Collapse
Affiliation(s)
- Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510280, China
- School of Dentistry, University of California Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
18
|
Sun J, Chang J, Guo Z, Sun H, Xu J, Liu X, Sun W. Proteomics Analysis of Renal Cell Line Caki-2 with AFMID Overexpression and Potential Biomarker Discovery in Urine. J Proteome Res 2024; 23:4495-4507. [PMID: 39213636 DOI: 10.1021/acs.jproteome.4c00431] [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: 09/04/2024]
Abstract
Aromatic caninurine formamase (AFMID) is an enzyme involved in the tryptophan pathway, metabolizing N-formylkynurenine to kynurenine. AFMID had been found significantly downregulated in clear cell renal cell carcinoma (ccRCC) in both tissue and urine samples. Although ccRCC is characterized by a typical Warburg-like phenotype, mitochondrial dysfunction, and elevated fat deposition, it is unknown whether AFMID plays a role in tumorigenesis and the development of ccRCC. In the present study, AFMID overexpression had inhibitory effects for ccRCC cells, decreasing the rate of cell proliferation. Quantitative proteomics showed that AFMID overexpression altered cellular signaling pathways involved in cell growth and cellular metabolism pathways, including lipid metabolism and inositol phosphate metabolism. Further urine proteomic analysis indicated that cellular function dysfunction with AFMID overexpression could be reflected in the urine. The activity of predicted upregulators DDX58, TREX1, TGFB1, SMARCA4, and TNF in ccRCC cells and urine showed opposing change trends. Potential urinary biomarkers were tentatively discovered and further validated using an independent cohort. The protein panel of APOC3, UMOD, and CILP achieved an AUC value of 0.862 for the training cohort and 0.883 for the validation cohort. The present study is of significance in terms of highlighting various aspects of pathway changes associated with AFMID enzymes, discovering potential specific biomarkers for potential patient diagnosis, and therapeutic targeting.
Collapse
Affiliation(s)
- Jiameng Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Jinchun Chang
- National Institute of Biological Sciences,7 Science Park Road ZGC Life Science Park, Beijing 102206, China
- School of Health, Quanzhou Medical College, No. 2 Anji Road, Luojiang District, Quanzhou City, Fujian Province 362011, China
| | - Zhengguang Guo
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Haidan Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Jiyu Xu
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xiaoyan Liu
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Wei Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| |
Collapse
|
19
|
Gulucu S, Onal M, Karakus N. Genetic Polymorphisms of COL1A1 Promoter Region (rs1800012) and TGFB1 Signal Peptide (rs1800471): Role in Cervical Insufficiency Susceptibility? Reprod Sci 2024; 31:3058-3065. [PMID: 39210235 DOI: 10.1007/s43032-024-01684-8] [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: 06/06/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
A structural or functional cervix problem prevents a woman from carrying a full-term pregnancy, which leads to the disease known as cervical insufficiency. Cervical insufficiency is partially inherited, and in certain situations, variations in genes related to connective tissue metabolism may be involved. The main objective of this investigation was to describe the collagen type I alpha 1 chain (COL1A1) gene rs1800012 polymorphism and the transforming growth factor beta 1 (TGFB1) gene rs1800471 polymorphism in a cohort of patients suffering from cervical insufficiency. Polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP) assays have been used to analyze the DNAs of 93 patients with cervical insufficiency and 103 healthy controls. The chi-square test was used for statistical analysis. There were significant differences in the genotype frequencies of the COL1A1 gene rs1800012 (G > T) and TGFB1 gene rs1800471 (G > C) polymorphisms between the patient and the control groups (p = 0.049 and p = 0.049, respectively). Also, the C allele of the TGFB1 rs1800471 polymorphism was significantly higher in the patient group than the control group (p = 0.016). Following clinical assessment, the COL1A1 rs1800012 polymorphism was found to be connected to the history of cerclage (p = 0.010). Additionally, the frequency of the TT/GG composite genotype of COL1A1 rs1800012/TGFB1 rs1800471 polymorphisms was significantly lower in the patient group than the control group (p = 0.049). The TT genotype of COL1A1 rs1800012 polymorphism was found to be protective against cervical insufficiency, while the C allele of TGFB1 rs1800471 polymorphism was found to predispose to the disease. It appears that the TT/GG composite genotype of COL1A1 rs1800012/TGFB1 rs1800471 polymorphisms protects against cervical insufficiency.
Collapse
Affiliation(s)
- Selim Gulucu
- Department of Gynecology and Obstetrics, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Turkey
| | - Mesut Onal
- Department of Gynecology and Obstetrics, Faculty of Medicine, Ondokuz Mayis University, Tokat, Turkey
| | - Nevin Karakus
- Department of Medical Biology, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Turkey.
| |
Collapse
|
20
|
Liu W, He H, Chicco D. Gene signatures for cancer research: A 25-year retrospective and future avenues. PLoS Comput Biol 2024; 20:e1012512. [PMID: 39413055 PMCID: PMC11482671 DOI: 10.1371/journal.pcbi.1012512] [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] [Indexed: 10/18/2024] Open
Abstract
Over the past two decades, extensive studies, particularly in cancer analysis through large datasets like The Cancer Genome Atlas (TCGA), have aimed at improving patient therapies and precision medicine. However, limited overlap and inconsistencies among gene signatures across different cohorts pose challenges. The dynamic nature of the transcriptome, encompassing diverse RNA species and functional complexities at gene and isoform levels, introduces intricacies, and current gene signatures face reproducibility issues due to the unique transcriptomic landscape of each patient. In this context, discrepancies arising from diverse sequencing technologies, data analysis algorithms, and software tools further hinder consistency. While careful experimental design, analytical strategies, and standardized protocols could enhance reproducibility, future prospects lie in multiomics data integration, machine learning techniques, open science practices, and collaborative efforts. Standardized metrics, quality control measures, and advancements in single-cell RNA-seq will contribute to unbiased gene signature identification. In this perspective article, we outline some thoughts and insights addressing challenges, standardized practices, and advanced methodologies enhancing the reliability of gene signatures in disease transcriptomic research.
Collapse
Affiliation(s)
- Wei Liu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Huaqin He
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Davide Chicco
- Dipartimento di Informatica Sistemistica e Comunicazione, Università di Milano-Bicocca, Milan, Italy
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Jiang YL, Li X, Tan YW, Fang YJ, Liu KY, Wang YF, Ma T, Ou QJ, Zhang CX. Docosahexaenoic acid inhibits the invasion and migration of colorectal cancer by reversing EMT through the TGF-β1/Smad signaling pathway. Food Funct 2024; 15:9420-9433. [PMID: 39189524 DOI: 10.1039/d4fo02346c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The primary cause of mortality in colorectal cancer (CRC) patients is tumor metastasis. The epithelial-mesenchymal transition (EMT) stands out as a crucial factor promoting the metastasis of CRC. Previous findings suggest a potential inhibitory effect of docosahexaenoic acid (DHA) on CRC metastasis, but the precise mechanism remains unknown, this study aims to explore this issue. We assessed metastasis and recurrence, all-cause mortality, and cancer-related mortality rates according to DHA intake in independent CRC cohorts (n = 367) by survival analysis. The ability of DHA to block CRC cell migration and invasion was tested using transwell and wound-healing assays. The regulation of EMT marker genes in CRC by DHA was detected by quantitative real-time PCR (qPCR) and immunoblotting, and the effect of DHA on the TGF-β1/Smad signaling pathway was further investigated. These cellular findings were validated using a subcutaneous CRC mouse model. Survival analyses showed that lower DHA intake was associated with a higher risk of CRC metastasis and a poorer prognosis. In vitro experiments showed that DHA inhibits the TGF-β1/Smad signaling pathway and regulates downstream transcription factors, thereby reversing the EMT and inhibiting invasion and migration. In the mouse model, dietary DHA supplementation effectively increased blood DHA concentrations and inhibited CRC metastasis. Our study demonstrated that DHA inhibits CRC invasion and metastasis by inhibiting the TGF-β1/Smad signaling pathway. Increased intake of DHA among CRC patients may provide additional benefits to the prognosis of colorectal cancer.
Collapse
Affiliation(s)
- Yi-Ling Jiang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Xue Li
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ya-Wen Tan
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yu-Jing Fang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai-Yan Liu
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yi-Fan Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ting Ma
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Qing-Jian Ou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Cai-Xia Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
22
|
Tang L, Qiu H, Xu B, Su Y, Nyarige V, Li P, Chen H, Killham B, Liao J, Adam H, Yang A, Yu A, Jang M, Rubart M, Xie J, Zhu W. Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. Circ Res 2024; 135:777-798. [PMID: 39145385 PMCID: PMC11392624 DOI: 10.1161/circresaha.124.324608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.
Collapse
Affiliation(s)
- Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Huiliang Qiu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Yajuan Su
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Verah Nyarige
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Houjia Chen
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Brady Killham
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Henderson Adam
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Alexander Yu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (M.R.)
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| |
Collapse
|
23
|
Pornour M, Jeon HY, Ryu H, Khadka S, Xu R, Chen H, Hussain A, Lam HM, Zhuang Z, Oo HZ, Gleave M, Dong X, Wang Q, Barbieri C, Qi J. USP11 promotes prostate cancer progression by up-regulating AR and c-Myc activity. Proc Natl Acad Sci U S A 2024; 121:e2403331121. [PMID: 39052835 PMCID: PMC11295044 DOI: 10.1073/pnas.2403331121] [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] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Androgen receptor (AR) is a main driver for castration-resistant prostate cancer (CRPC). c-Myc is an oncogene underlying prostate tumorigenesis. Here, we find that the deubiquitinase USP11 targets both AR and c-Myc in prostate cancer (PCa). USP11 expression was up-regulated in metastatic PCa and CRPC. USP11 knockdown (KD) significantly inhibited PCa cell growth. Our RNA-seq studies revealed AR and c-Myc as the top transcription factors altered after USP11 KD. ChIP-seq analysis showed that either USP11 KD or replacement of endogenous USP11 with a catalytic-inactive USP11 mutant significantly decreased chromatin binding by AR and c-Myc. We find that USP11 employs two mechanisms to up-regulate AR and c-Myc levels: namely, deubiquitination of AR and c-Myc proteins to increase their stability and deubiquitination of H2A-K119Ub, a repressive histone mark, on promoters of AR and c-Myc genes to increase their transcription. AR and c-Myc reexpression in USP11-KD PCa cells partly rescued cell growth defects. Thus, our studies reveal a tumor-promoting role for USP11 in aggressive PCa through upregulation of AR and c-Myc activities and support USP11 as a potential target against PCa.
Collapse
Affiliation(s)
- Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Hee-Young Jeon
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Hyunju Ryu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Sudeep Khadka
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| | - Rui Xu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland, Baltimore, MD21202
| | - Hegang Chen
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, MD21201
| | - Arif Hussain
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
- Baltimore Veterans Affairs Medical Center, Baltimore, MD21201
| | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, WA98195
| | - Zhihao Zhuang
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE19716
| | - Htoo Zarni Oo
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
| | - Martin Gleave
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
| | - Xuesen Dong
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
| | - Qianben Wang
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, NC27710
| | - Christopher Barbieri
- Department of Urology, Weill Cornell Medical College, Cornell University, New York, NY10065
| | - Jianfei Qi
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD21201
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD21201
| |
Collapse
|
24
|
Fathima A, Farboodniay Jahromi MA, Begum SA, Jamma T. Withametelin inhibits TGF-β induced Epithelial-to-Mesenchymal Transition and Programmed-Death Ligand-1 expression in vitro. Front Oncol 2024; 14:1435516. [PMID: 39077463 PMCID: PMC11284055 DOI: 10.3389/fonc.2024.1435516] [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: 05/20/2024] [Accepted: 06/26/2024] [Indexed: 07/31/2024] Open
Abstract
Withanolides are a group of naturally occurring plant-based small molecules known for their wide range of host cellular functions. The anticancer potential of withanolides has been explored in varying cancer cell lines in vitro. Based on our prior studies, among the tested withanolides, withametelin (WM) has shown significant cytotoxicity with the highest efficacy on HCT-116 colon cancer cells (IC50 0.719 ± 0.12μM). Treatment with WM reduced the TGF-β driven proliferation, colony-forming ability, migration, and invasiveness of HCT-116 cells in vitro. WM also downregulated the expression of mesenchymal markers such as N-CADHERIN, SNAIL, and SLUG in HCT-116 cells. At the molecular level, WM inhibited TGF-β induced phosphorylation of SMAD2/3 and reduced the expression of an immune checkpoint inhibitor programmed-death ligand-1 (PD-L1). Our study highlights the possible anticancer mechanisms of WM involving modulation of the TGF-β pathway and associated target gene expression, suggesting its potential utility in cancer therapy.
Collapse
Affiliation(s)
- Ashna Fathima
- Cell Signaling Laboratory, Department of Biological Sciences, Birla Institute of Technology & Science, Hyderabad, India
| | | | - Sajeli A. Begum
- Department of Pharmacy, Birla Institute of Technology & Science, Hyderabad, India
| | - Trinath Jamma
- Cell Signaling Laboratory, Department of Biological Sciences, Birla Institute of Technology & Science, Hyderabad, India
| |
Collapse
|
25
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [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: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
26
|
Louault K, Blavier L, Lee MH, Kennedy RJ, Fernandez GE, Pawel BR, Asgharzadeh S, DeClerck YA. Nuclear factor-κB activation by transforming growth factor-β1 drives tumour microenvironment-mediated drug resistance in neuroblastoma. Br J Cancer 2024; 131:90-100. [PMID: 38806726 PMCID: PMC11231159 DOI: 10.1038/s41416-024-02686-8] [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: 10/13/2023] [Revised: 03/26/2024] [Accepted: 04/08/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Intrinsic and extrinsic factors in the tumour microenvironment (TME) contribute to therapeutic resistance. Here we demonstrate that transforming growth factor (TGF)-β1 produced in the TME increased drug resistance of neuroblastoma (NB) cells. METHODS Human NB cell lines were tested in vitro for their sensitivity to Doxorubicin (DOX) and Etoposide (ETOP) in the presence of tumour-associated macrophages (TAM) and mesenchymal stromal cells/cancer-associated fibroblasts (MSC/CAF). These experiments were validated in xenotransplanted and primary tumour samples. RESULTS Drug resistance was associated with an increased expression of efflux transporter and anti-apoptotic proteins. Upregulation was dependent on activation of nuclear factor (NF)-κB by TGF-β-activated kinase (TAK1) and SMAD2. Resistance was reversed upon pharmacologic and genetic inhibitions of NF-κB, and TAK1/SMAD2. Interleukin-6, leukaemia inhibitory factor and oncostatin M were upregulated by this TGF-β/TAK1/NF-κB/SMAD2 signalling pathway contributing to drug resistance via an autocrine loop activating STAT3. An analysis of xenotransplanted NB tumours revealed an increased presence of phospho (p)-NF-κB in tumours co-injected with MSC/CAF and TAM, and these tumours failed to respond to Etoposide but responded if treated with a TGF-βR1/ALK5 inhibitor. Nuclear p-NF-κB was increased in patient-derived tumours rich in TME cells. CONCLUSIONS The data provides a novel insight into a targetable mechanism of environment-mediated drug resistance.
Collapse
Affiliation(s)
- Kévin Louault
- Cancer and Blood Diseases Institute, Department of Pediatrics, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, 90027, USA
| | - Laurence Blavier
- Cancer and Blood Diseases Institute, Department of Pediatrics, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, 90027, USA
| | - Men-Hua Lee
- Cancer and Blood Diseases Institute, Department of Pediatrics, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, 90027, USA
| | - Rebekah J Kennedy
- Cancer and Blood Diseases Institute, Department of Pediatrics, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, 90027, USA
| | - G Esteban Fernandez
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Bruce R Pawel
- Department of Pathology, and Laboratory Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shahab Asgharzadeh
- Cancer and Blood Diseases Institute, Department of Pediatrics, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, 90027, USA
- Department of Pathology, and Laboratory Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Yves A DeClerck
- Cancer and Blood Diseases Institute, Department of Pediatrics, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, 90027, USA.
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
27
|
Li X, Wang Y, Wang X, Shen Y, Yuan Y, He Q, Mao S, Wu C, Zhou M. Downregulation of SMAD4 protects HaCaT cells against UVB-induced damage and oxidative stress through the activation of EMT. Photochem Photobiol Sci 2024; 23:1051-1065. [PMID: 38684635 DOI: 10.1007/s43630-024-00574-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/04/2024] [Indexed: 05/02/2024]
Abstract
As a member of the SMAD family, SMAD4 plays a crucial role in several cellular biological processes. However, its function in UVB radiation-induced keratinocyte damage is not yet clarified. Our study aims to provide mechanistic insight for the development of future UVB protective therapies and therapeutics involving SMAD4. HaCaT cells were treated with UVB, and the dose dependence and time dependence of UVB were measured. The cell function of UVB-treated HaCaT cells and the activity of epithelial-mesenchymal transition (EMT) after overexpression or silencing of SMAD4 was observed by flow cytometry, quantitative reverse transcription PCR (qRT-PCR) and Western Blots (WB). We found that a significant decrease in SMAD4 was observed in HaCaT cells induced by UVB. Our data confirm SMAD4 as a direct downstream target of miR-664. The down-regulation of SMAD4 preserved the viability of the UVB-treated HaCaT cells by inhibiting autophagy or apoptosis. Furthermore, the silencing of SMAD4 activated the EMT process in UVB-treated HaCaT cells. Down-regulation of SMAD4 plays a protective role in UVB-treated HaCaT cells via the activation of EMT.
Collapse
Affiliation(s)
- Xiangzhi Li
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545000, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, 317502, China
| | - Yimeng Wang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
- Yancheng Center for Disease Control and Prevention, Yancheng, 224000, China
| | - Xian Wang
- Department of Public Health and Management, Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yi Shen
- Department of Public Health and Management, Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yawen Yuan
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Qingquan He
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Branch of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou, 317502, China
| | - Shuyi Mao
- Nuclear Medicine Department, The Second Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, 545006, China
| | - Cailian Wu
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545000, China
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
28
|
Guo X, Fu Y, Peng J, Fu Y, Dong S, Ding RB, Qi X, Bao J. Emerging anticancer potential and mechanisms of snake venom toxins: A review. Int J Biol Macromol 2024; 269:131990. [PMID: 38704067 DOI: 10.1016/j.ijbiomac.2024.131990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/13/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Animal-derived venom, like snake venom, has been proven to be valuable natural resources for the drug development. Previously, snake venom was mainly investigated in its pharmacological activities in regulating coagulation, vasodilation, and cardiovascular function, and several marketed cardiovascular drugs were successfully developed from snake venom. In recent years, snake venom fractions have been demonstrated with anticancer properties of inducing apoptotic and autophagic cell death, restraining proliferation, suppressing angiogenesis, inhibiting cell adhesion and migration, improving immunity, and so on. A number of active anticancer enzymes and peptides have been identified from snake venom toxins, such as L-amino acid oxidases (LAAOs), phospholipase A2 (PLA2), metalloproteinases (MPs), three-finger toxins (3FTxs), serine proteinases (SPs), disintegrins, C-type lectin-like proteins (CTLPs), cell-penetrating peptides, cysteine-rich secretory proteins (CRISPs). In this review, we focus on summarizing these snake venom-derived anticancer components on their anticancer activities and underlying mechanisms. We will also discuss their potential to be developed as anticancer drugs in the future.
Collapse
Affiliation(s)
- Xijun Guo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanfeng Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Junbo Peng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ying Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Shuai Dong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xingzhu Qi
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China.
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
29
|
Ciccarelli EJ, Wing Z, Bendelstein M, Johal RK, Singh G, Monas A, Savage-Dunn C. TGF-β ligand cross-subfamily interactions in the response of Caenorhabditis elegans to a bacterial pathogen. PLoS Genet 2024; 20:e1011324. [PMID: 38875298 PMCID: PMC11210861 DOI: 10.1371/journal.pgen.1011324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 06/27/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
The Transforming Growth Factor beta (TGF-β) family consists of numerous secreted peptide growth factors that play significant roles in cell function, tissue patterning, and organismal homeostasis, including wound repair and immunity. Typically studied as homodimers, these ligands have the potential to diversify their functions through ligand interactions that may enhance, repress, or generate novel functions. In the nematode Caenorhabditis elegans, there are only five TGF-β ligands, providing an opportunity to dissect ligand interactions in fewer combinations than in vertebrates. As in vertebrates, these ligands can be divided into bone morphogenetic protein (BMP) and TGF-β/Activin subfamilies that predominantly signal through discrete signaling pathways. The BMP subfamily ligand DBL-1 has been well studied for its role in the innate immune response in C. elegans. Here we show that all five TGF-β ligands play a role in survival on bacterial pathogens. We also demonstrate that multiple TGF-β ligand pairs act nonredundantly as part of this response. We show that the two BMP-like ligands-DBL-1 and TIG-2-function independently of each other in the immune response, while TIG-2/BMP and the TGF-β/Activin-like ligand TIG-3 function together. Structural modeling supports the potential for TIG-2 and TIG-3 to form heterodimers. Additionally, we identify TIG-2 and TIG-3 as members of a rare subset of TGF-β ligands lacking the conserved cysteine responsible for disulfide linking mature dimers. Finally, we show that canonical DBL-1/BMP receptor and Smad signal transducers function in the response to bacterial pathogens, while components of the DAF-7 TGF-β/Activin signaling pathway do not play a major role in survival. These results demonstrate a novel potential for BMP and TGF-β/Activin subfamily ligands to interact and may provide a mechanism for distinguishing the developmental and homeostatic functions of these ligands from an acute response such as the innate immune response to bacterial pathogens.
Collapse
Affiliation(s)
- Emma Jo Ciccarelli
- Department of Biology, Queens College, City University of New York, New York City, New York, United States of America
- PhD Program in Biology, The Graduate Center, City University of New York, New York City, New York, United States of America
| | - Zachary Wing
- Department of Biology, Queens College, City University of New York, New York City, New York, United States of America
| | - Moshe Bendelstein
- Department of Biology, Queens College, City University of New York, New York City, New York, United States of America
| | - Ramandeep Kaur Johal
- Department of Biology, Queens College, City University of New York, New York City, New York, United States of America
| | - Gurjot Singh
- Department of Biology, Queens College, City University of New York, New York City, New York, United States of America
| | - Ayelet Monas
- Department of Biology, Queens College, City University of New York, New York City, New York, United States of America
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, City University of New York, New York City, New York, United States of America
- PhD Program in Biology, The Graduate Center, City University of New York, New York City, New York, United States of America
| |
Collapse
|
30
|
Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys 2024; 119:42-55. [PMID: 38042450 PMCID: PMC11082936 DOI: 10.1016/j.ijrobp.2023.11.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Radiation therapy (RT) has been a primary treatment modality in cancer for decades. Increasing evidence suggests that RT can induce an immunosuppressive shift via upregulation of cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). MDSCs inhibit antitumor immunity through potent immunosuppressive mechanisms and have the potential to be crucial tools for cancer prognosis and treatment. MDSCs interact with many different pathways, desensitizing tumor tissue and interacting with tumor cells to promote therapeutic resistance. Vascular damage induced by RT triggers an inflammatory signaling cascade and potentiates hypoxia in the tumor microenvironment (TME). RT can also drastically modify cytokine and chemokine signaling in the TME to promote the accumulation of MDSCs. RT activation of the cGAS-STING cytosolic DNA sensing pathway recruits MDSCs through a CCR2-mediated mechanism, inhibiting the production of type 1 interferons and hampering antitumor activity and immune surveillance in the TME. The upregulation of hypoxia-inducible factor-1 and vascular endothelial growth factor mobilizes MDSCs to the TME. After recruitment, MDSCs promote immunosuppression by releasing reactive oxygen species and upregulating nitric oxide production through inducible nitric oxide synthase expression to inhibit cytotoxic activity. Overexpression of arginase-1 on subsets of MDSCs degrades L-arginine and downregulates CD3ζ, inhibiting T-cell receptor reactivity. This review explains how radiation promotes tumor resistance through activation of immunosuppressive MDSCs in the TME and discusses current research targeting MDSCs, which could serve as a promising clinical treatment strategy in the future.
Collapse
Affiliation(s)
| | - Matthew Berkseth
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sudipto Ganguly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | | | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
31
|
Yu P, Han Y, Meng L, Tang Z, Jin Z, Zhang Z, Zhou Y, Luo J, Luo J, Han C, Zhang C, Kong L. The incorporation of acetylated LAP-TGF-β1 proteins into exosomes promotes TNBC cell dissemination in lung micro-metastasis. Mol Cancer 2024; 23:82. [PMID: 38664722 PMCID: PMC11044330 DOI: 10.1186/s12943-024-01995-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
Triple-negative breast cancer (TNBC) stands as the breast cancer subtype with the highest recurrence and mortality rates, with the lungs being the common site of metastasis. The pulmonary microenvironment plays a pivotal role in the colonization of disseminated tumor cells. Herein, this study highlights the crucial role of exosomal LAP-TGF-β1, the principal form of exosomal TGF-β1, in reshaping the pulmonary vascular niche, thereby facilitating TNBC lung metastasis. Although various strategies have been developed to block TGF-β signaling and have advanced clinically, their significant side effects have limited their therapeutic application. This study demonstrates that in lung metastatic sites, LAP-TGF-β1 within exosomes can remarkably reconfigure the pulmonary vascular niche at lower doses, bolstering the extravasation and colonization of TNBC cells in the lungs. Mechanistically, under the aegis of the acetyltransferase TIP60, a non-canonical KFERQ-like sequence in LAP-TGF-β1 undergoes acetylation at the K304 site, promoting its interaction with HSP90A and subsequent transport into exosomes. Concurrent inhibition of both HSP90A and TIP60 significantly diminishes the exosomal burden of LAP-TGF-β1, presenting a promising therapeutic avenue for TNBC lung metastasis. This study not only offers fresh insights into the molecular underpinnings of TNBC lung metastasis but also lays a foundation for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Pei Yu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yubao Han
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Lulu Meng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Zengying Tang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhiwei Jin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhenzhen Zhang
- Institute of Veterinary Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Yunjiang Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jun Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jianguang Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chao Han
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chao Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
32
|
Danielpour D. Advances and Challenges in Targeting TGF-β Isoforms for Therapeutic Intervention of Cancer: A Mechanism-Based Perspective. Pharmaceuticals (Basel) 2024; 17:533. [PMID: 38675493 PMCID: PMC11054419 DOI: 10.3390/ph17040533] [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: 02/27/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The TGF-β family is a group of 25 kDa secretory cytokines, in mammals consisting of three dimeric isoforms (TGF-βs 1, 2, and 3), each encoded on a separate gene with unique regulatory elements. Each isoform plays unique, diverse, and pivotal roles in cell growth, survival, immune response, and differentiation. However, many researchers in the TGF-β field often mistakenly assume a uniform functionality among all three isoforms. Although TGF-βs are essential for normal development and many cellular and physiological processes, their dysregulated expression contributes significantly to various diseases. Notably, they drive conditions like fibrosis and tumor metastasis/progression. To counter these pathologies, extensive efforts have been directed towards targeting TGF-βs, resulting in the development of a range of TGF-β inhibitors. Despite some clinical success, these agents have yet to reach their full potential in the treatment of cancers. A significant challenge rests in effectively targeting TGF-βs' pathological functions while preserving their physiological roles. Many existing approaches collectively target all three isoforms, failing to target just the specific deregulated ones. Additionally, most strategies tackle the entire TGF-β signaling pathway instead of focusing on disease-specific components or preferentially targeting tumors. This review gives a unique historical overview of the TGF-β field often missed in other reviews and provides a current landscape of TGF-β research, emphasizing isoform-specific functions and disease implications. The review then delves into ongoing therapeutic strategies in cancer, stressing the need for more tools that target specific isoforms and disease-related pathway components, advocating mechanism-based and refined approaches to enhance the effectiveness of TGF-β-targeted cancer therapies.
Collapse
Affiliation(s)
- David Danielpour
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology, Case Western Reserve University, Cleveland, OH 44106, USA; ; Tel.: +1-216-368-5670; Fax: +1-216-368-8919
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
- Institute of Urology, University Hospitals, Cleveland, OH 44106, USA
| |
Collapse
|
33
|
Reshkin SJ, Cardone RA, Koltai T. Genetic Signature of Human Pancreatic Cancer and Personalized Targeting. Cells 2024; 13:602. [PMID: 38607041 PMCID: PMC11011857 DOI: 10.3390/cells13070602] [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: 02/12/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Pancreatic cancer is a highly lethal disease with a 5-year survival rate of around 11-12%. Surgery, being the treatment of choice, is only possible in 20% of symptomatic patients. The main reason is that when it becomes symptomatic, IT IS the tumor is usually locally advanced and/or has metastasized to distant organs; thus, early diagnosis is infrequent. The lack of specific early symptoms is an important cause of late diagnosis. Unfortunately, diagnostic tumor markers become positive at a late stage, and there is a lack of early-stage markers. Surgical and non-surgical cases are treated with neoadjuvant and/or adjuvant chemotherapy, and the results are usually poor. However, personalized targeted therapy directed against tumor drivers may improve this situation. Until recently, many pancreatic tumor driver genes/proteins were considered untargetable. Chemical and physical characteristics of mutated KRAS are a formidable challenge to overcome. This situation is slowly changing. For the first time, there are candidate drugs that can target the main driver gene of pancreatic cancer: KRAS. Indeed, KRAS inhibition has been clinically achieved in lung cancer and, at the pre-clinical level, in pancreatic cancer as well. This will probably change the very poor outlook for this disease. This paper reviews the genetic characteristics of sporadic and hereditary predisposition to pancreatic cancer and the possibilities of a personalized treatment according to the genetic signature.
Collapse
Affiliation(s)
- Stephan J. Reshkin
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Tomas Koltai
- Oncomed, Via Pier Capponi 6, 50132 Florence, Italy
| |
Collapse
|
34
|
Saadh MJ, Allela OQB, Sattay ZJ, Al Zuhairi RAH, Ahmad H, Eldesoky GE, Adil M, Ali MS. Deciphering the functional landscape and therapeutic implications of noncoding RNAs in the TGF-β signaling pathway in colorectal cancer: A comprehensive review. Pathol Res Pract 2024; 255:155158. [PMID: 38320438 DOI: 10.1016/j.prp.2024.155158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/08/2024]
Abstract
Colorectal cancer (CRC) remains a major global health concern, necessitating an in-depth exploration of the intricate molecular mechanisms underlying its progression and potential therapeutic interventions. Transforming Growth Factor-β (TGF-β) signaling, a pivotal pathway implicated in CRC plays a dual role as a tumor suppressor in the early stages and a promoter of tumor progression in later stages. Recent research has shed light on the critical involvement of noncoding RNAs (ncRNAs) in modulating the TGF-β signaling pathway, introducing a new layer of complexity to our understanding of CRC pathogenesis. This comprehensive review synthesizes the current state of knowledge regarding the function and therapeutic potential of various classes of ncRNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), in the context of TGF-β signaling in CRC. The intricate interplay between these ncRNAs and key components of the TGF-β pathway is dissected, revealing regulatory networks that contribute to the dynamic balance between tumor suppression and promotion. Emphasis is placed on how dysregulation of specific ncRNAs can disrupt this delicate equilibrium, fostering CRC initiation, progression, and metastasis. Moreover, the review provides a critical appraisal of the emerging therapeutic strategies targeting ncRNAs associated with TGF-β signaling in CRC. The potential of these ncRNAs as diagnostic and prognostic biomarkers is discussed, highlighting their clinical relevance. Additionally, the challenges and prospects of developing RNA-based therapeutics, such as RNA interference and CRISPR/Cas-based approaches, are explored in the context of modulating TGF-β signaling for CRC treatment. In conclusion, this review offers a comprehensive overview of the intricate interplay between ncRNAs and the TGF-β signaling pathway in CRC. By unraveling the functional significance of these regulatory elements, we gain valuable insights into the molecular landscape of CRC, paving the way for the development of novel and targeted therapeutic interventions aimed at modulating the TGF-β signaling cascade through the manipulation of ncRNAs.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | - Zahraa Jasim Sattay
- Department of Medical Laboratory Technology l, University of imam Jaafar Al-Sadiq, Iraq
| | | | - Hijaz Ahmad
- Section of Mathematics, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, Rome 00186, Italy; Center for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Kuwait; Department of Computer Science and Mathematics, Lebanese American University, Beirut, Lebanon
| | - Gaber E Eldesoky
- Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | | | | |
Collapse
|
35
|
Sacco JL, Vaneman ZT, Gomez EW. Extracellular matrix viscoelasticity regulates TGFβ1-induced epithelial-mesenchymal transition and apoptosis via integrin linked kinase. J Cell Physiol 2024; 239:e31165. [PMID: 38149820 DOI: 10.1002/jcp.31165] [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: 06/21/2023] [Revised: 10/06/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023]
Abstract
Transforming growth factor (TGF)-β1 is a multifunctional cytokine that plays important roles in health and disease. Previous studies have revealed that TGFβ1 activation, signaling, and downstream cell responses including epithelial-mesenchymal transition (EMT) and apoptosis are regulated by the elasticity or stiffness of the extracellular matrix. However, tissues within the body are not purely elastic, rather they are viscoelastic. How matrix viscoelasticity impacts cell fate decisions downstream of TGFβ1 remains unknown. Here, we synthesized polyacrylamide hydrogels that mimic the viscoelastic properties of breast tumor tissue. We found that increasing matrix viscous dissipation reduces TGFβ1-induced cell spreading, F-actin stress fiber formation, and EMT-associated gene expression changes, and promotes TGFβ1-induced apoptosis in mammary epithelial cells. Furthermore, TGFβ1-induced expression of integrin linked kinase (ILK) and colocalization of ILK with vinculin at cell adhesions is attenuated in mammary epithelial cells cultured on viscoelastic substrata in comparison to cells cultured on nearly elastic substrata. Overexpression of ILK promotes TGFβ1-induced EMT and reduces apoptosis in cells cultured on viscoelastic substrata, suggesting that ILK plays an important role in regulating cell fate downstream of TGFβ1 in response to matrix viscoelasticity.
Collapse
Affiliation(s)
- Jessica L Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Zachary T Vaneman
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Esther W Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
36
|
Ahuja S, Zaheer S. Multifaceted TGF-β signaling, a master regulator: From bench-to-bedside, intricacies, and complexities. Cell Biol Int 2024; 48:87-127. [PMID: 37859532 DOI: 10.1002/cbin.12097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Physiological embryogenesis and adult tissue homeostasis are regulated by transforming growth factor-β (TGF-β), an evolutionarily conserved family of secreted polypeptide factors, acting in an autocrine and paracrine manner. The role of TGF-β in inflammation, fibrosis, and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, especially fibrosis and cancer, overexpressed TGF-β causes extracellular matrix deposition, epithelial-mesenchymal transition, cancer-associated fibroblast formation, and/or angiogenesis. In this review article, we have tried to dive deep into the mechanism of action of TGF-β in inflammation, fibrosis, and carcinogenesis. As TGF-β and its downstream signaling mechanism are implicated in fibrosis and carcinogenesis blocking this signaling mechanism appears to be a promising avenue. However, targeting TGF-β carries substantial risk as this pathway is implicated in multiple homeostatic processes and is also known to have tumor-suppressor functions. There is a need for careful dosing of TGF-β drugs for therapeutic use and patient selection.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
37
|
Szilágyi SS, Burdzinski W, Jatzlau J, Ehrlich M, Knaus P, Henis YI. The Activation of the Fibrodysplasia Ossificans Progressiva-Inducing ALK2-R206H Mutant Depends on the Distinct Homo-Oligomerization Patterns of ACVR2B and ACVR2A. Cells 2024; 13:221. [PMID: 38334613 PMCID: PMC10854824 DOI: 10.3390/cells13030221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Mutations in activin-like kinase 2 (ALK2), e.g., ALK2-R206H, induce aberrant signaling to SMAD1/5/8, leading to Fibrodysplasia Ossificans Progressiva (FOP). In spite of extensive studies, the underlying mechanism is still unclear. Here, we quantified the homomeric and heteromeric interactions of ACVR2A, ACVR2B, ALK2-WT, and ALK2-R206H by combining IgG-mediated immobilization of one receptor with fluorescence recovery after photobleaching (FRAP) measurements on the lateral diffusion of a co-expressed receptor. ACVR2B formed stable homomeric complexes that were enhanced by Activin A (ActA), while ACVR2A required ActA for homodimerization. ALK2-WT, but not ALK2-R206H, exhibited homomeric complexes unaffected by ActA. ACVR2B formed ActA-enhanced heterocomplexes with ALK2-R206H or ALK2-WT, while ACVR2A interacted mainly with ALK2-WT. The extent of the homomeric complex formation of ACVR2A or ACVR2B was reflected in their ability to induce the oligomerization of ALK2-R206H and ALK2-WT. Thus, ACVR2B, which forms dimers without ligand, induced ActA-independent ALK2-R206H clustering but required ActA for enhancing the oligomerization of the largely dimeric ALK2-WT. In contrast, ACVR2A, which undergoes homodimerization in response to ActA, required ActA to induce ALK2-R206H oligomerization. To investigate whether these interactions are translated into signaling, we studied signaling by the FOP-inducing hyperactive ALK2-R206H mutant, with ALK2-WT signaling as control. The activation of SMAD1/5/8 signaling in cells expressing ALK2-R206H alone or together with ACVR2A or ACVR2B was measured by blotting for pSMAD1/5/8 and by transcriptional activation assays using BRE-Luc reporter. In line with the biophysical studies, ACVR2B activated ALK2-R206H without ligand, while activation by ACVR2A was weaker and required ActA. We propose that the homodimerization of ACVR2B or ACVR2A dictates their ability to recruit ALK2-R206H into higher complexes, enabling the homomeric interactions of ALK2-R206H receptors and, subsequently, their activation.
Collapse
Affiliation(s)
- Szabina Szófia Szilágyi
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Wiktor Burdzinski
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jerome Jatzlau
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany (J.J.); (P.K.)
| | - Yoav I. Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| |
Collapse
|
38
|
Wang D, Nakayama M, Hong CP, Oshima H, Oshima M. Gain-of-Function p53 Mutation Acts as a Genetic Switch for TGFβ Signaling-Induced Epithelial-to-Mesenchymal Transition in Intestinal Tumors. Cancer Res 2024; 84:56-68. [PMID: 37851521 PMCID: PMC10758690 DOI: 10.1158/0008-5472.can-23-1490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/08/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Signaling by TGFβ family cytokines plays a tumor-suppressive role by inducing cell differentiation, while it promotes malignant progression through epithelial-to-mesenchymal transition (EMT). Identification of the mechanisms regulating the switch from tumor suppression to tumor promotion could identify strategies for cancer prevention and treatment. To identify the key genetic alterations that determine the outcome of TGFβ signaling, we used mouse intestinal tumor-derived organoids carrying multiple driver mutations in various combinations to examine the relationship between genotypes and responses to the TGFβ family cytokine activin A. KrasG12D mutation protected organoid cells from activin A-induced growth suppression by inhibiting p21 and p27 expression. Furthermore, Trp53R270H gain-of-function (GOF) mutation together with loss of wild-type Trp53 by loss of heterozygosity (LOH) promoted activin A-induced partial EMT with formation of multiple protrusions on the organoid surface, which was associated with increased metastatic incidence. Histologic analysis confirmed that tumor cells at the protrusions showed loss of apical-basal polarity and glandular structure. RNA sequencing analysis indicated that expression of Hmga2, encoding a cofactor of the SMAD complex that induces EMT transcription factors, was significantly upregulated in organoids with Trp53 GOF/LOH alterations. Importantly, loss of HMGA2 suppressed expression of Twist1 and blocked activin A-induced partial EMT and metastasis in Trp53 GOF/LOH organoids. These results indicate that TP53 GOF/LOH is a key genetic state that primes for TGFβ family-induced partial EMT and malignant progression of colorectal cancer. Activin signaling may be an effective therapeutic target for colorectal cancer harboring TP53 GOF mutations. SIGNIFICANCE KRAS and TP53 mutations shift activin-mediated signaling to overcome growth inhibition and promote partial EMT, identifying a subset of patients with colorectal cancer that could benefit from inhibition of TGFβ signaling.
Collapse
Affiliation(s)
- Dong Wang
- WPI Nano-Life Science Institute (Nano-LSI), Kanazawa University, Kanazawa, Japan
| | - Mizuho Nakayama
- WPI Nano-Life Science Institute (Nano-LSI), Kanazawa University, Kanazawa, Japan
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | - Hiroko Oshima
- WPI Nano-Life Science Institute (Nano-LSI), Kanazawa University, Kanazawa, Japan
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- WPI Nano-Life Science Institute (Nano-LSI), Kanazawa University, Kanazawa, Japan
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
39
|
He J, Wu F, Li J, Deng Q, Chen J, Li P, Jiang X, Yang K, Xu S, Jiang Z, Li X, Jiang Z. Tumor suppressor CLCA1 inhibits angiogenesis via TGFB1/SMAD/VEGF cascade and sensitizes hepatocellular carcinoma cells to Sorafenib. Dig Liver Dis 2024; 56:176-186. [PMID: 37230858 DOI: 10.1016/j.dld.2023.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/18/2023] [Accepted: 05/05/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly vascularized tumor with a poor prognosis. Novel vascular-related therapeutic targets and prognostic markers remain urgently needed. AIMS To investigate the role and mechanism of CLCA1 in hepatocellular carcinoma. METHODS Immunofluorescence, Co-immunoprecipitation and rescue experiment were used to determine the specific mechanisms of CLCA1. Chemosensitivity assay was used to measure the impact of CLCA1 on Sorafenib. RESULTS CLCA1 was dramatically downregulated in hepatocellular carcinoma cell lines and tissues. Ectopic expression of CLCA1 induced cell apoptosis and G0/G1 phase arrest while suppressed cell growth, inhibited migration and invasion, reversal of epithelial mesenchymal transition in vitro and reduced xenograft tumor growth in vivo. Mechanistically, CLCA1 could co-localize and interact with TGFB1, thereby suppressing HCC angiogenesis through the TGFB1/SMAD/VEGF signaling cascade in vitro and in vivo. Moreover, CLCA1 also enhanced the sensitivity of HCC cells to the first-line targeted therapy, Sorafenib. CONCLUSION CLCA1 sensitizes HCC cells to Sorafenib and suppresses hepatocellular carcinoma angiogenesis through downregulating TGFB1 signaling cascade. This newly identified CLCA1 signaling pathway may help guide the anti-angiogenesis therapies for hepatocellular carcinoma. We also support the possibility of CLCA1 being a prognostic biomarker for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Junfeng Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qianxi Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Pengtao Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Beijing 100044, China
| | - Xianyao Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital, Haikou 570100, China
| | - Kun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shuman Xu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhongxiang Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaoqing Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
40
|
Zhong S, Du X, Gao J, Ji G, Liu Z. BMP8B Activates Both SMAD2/3 and NF-κB Signals to Inhibit the Differentiation of 3T3-L1 Preadipocytes into Mature Adipocytes. Nutrients 2023; 16:64. [PMID: 38201894 PMCID: PMC10780770 DOI: 10.3390/nu16010064] [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: 11/14/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Bone morphogenetic protein 8B (BMP8B) has been found to regulate the thermogenesis of brown adipose tissue (BAT) and the browning process of white adipose tissue (WAT). However, there is no available information regarding the role of BMP8B in the process of adipocyte differentiation. Here, we showed that BMP8B down-regulates transcriptional regulators PPARγ and C/EBPα, thereby impeding the differentiation of 3T3-L1 preadipocytes into fully mature adipocytes. BMP8B increased the phosphorylation levels of SMAD2/3, and TP0427736 HCl (SMAD2/3 inhibitor) significantly reduced the ability of BMP8B to inhibit adipocyte differentiation, suggesting that BMP8B repressed adipocyte differentiation through the SMAD2/3 pathway. Moreover, the knockdown of BMP I receptor ALK4 significantly reduced the inhibitory effect of BMP8B on adipogenesis, indicating that BMP8B triggers SMAD2/3 signaling to suppress adipogenesis via ALK4. In addition, BMP8B activated the NF-κB signal, which has been demonstrated to impede PPARγ expression. Collectively, our data demonstrated that BMP8B activates both SMAD2/3 and NF-κB signals to inhibit adipocyte differentiation. We provide previously unidentified insight into BMP8B-mediated adipogenesis.
Collapse
Affiliation(s)
- Shenjie Zhong
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.Z.); (X.D.); (J.G.); (G.J.)
| | - Xueqing Du
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.Z.); (X.D.); (J.G.); (G.J.)
| | - Jing Gao
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.Z.); (X.D.); (J.G.); (G.J.)
| | - Guangdong Ji
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.Z.); (X.D.); (J.G.); (G.J.)
- Laoshan Laboratory, Qingdao 266237, China
| | - Zhenhui Liu
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.Z.); (X.D.); (J.G.); (G.J.)
- Laoshan Laboratory, Qingdao 266237, China
| |
Collapse
|
41
|
Nagasaka M, Inoue Y, Nagao Y, Miyajima C, Morishita D, Aoki H, Aoyama M, Imamura T, Hayashi H. SET8 is a novel negative regulator of TGF-β signaling in a methylation-independent manner. Sci Rep 2023; 13:22877. [PMID: 38129484 PMCID: PMC10739863 DOI: 10.1038/s41598-023-49961-x] [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: 09/25/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Transforming growth factor β (TGF-β) is a multifunctional cytokine that induces a diverse set of cellular processes principally through Smad-dependent transcription. Transcriptional responses induced by Smads are tightly regulated by Smad cofactors and histone modifications; however, the underlying mechanisms have not yet been elucidated in detail. We herein report lysine methyltransferase SET8 as a negative regulator of TGF-β signaling. SET8 physically associates with Smad2/3 and negatively affects transcriptional activation by TGF-β in a catalytic activity-independent manner. The depletion of SET8 results in an increase in TGF-β-induced plasminogen activator inhibitor-1 (PAI-1) and p21 expression and enhances the antiproliferative effects of TGF-β. Mechanistically, SET8 occupies the PAI-1 and p21 promoters, and a treatment with TGF-β triggers the replacement of the suppressive binding of SET8 with p300 on these promoters, possibly to promote gene transcription. Collectively, the present results reveal a novel role for SET8 in the negative regulation of TGF-β signaling.
Collapse
Affiliation(s)
- Mai Nagasaka
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Yasumichi Inoue
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| | - Yuji Nagao
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Chiharu Miyajima
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Daisuke Morishita
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Mineyoshi Aoyama
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| |
Collapse
|
42
|
Kondapuram SK, Ramachandran HK, Arya H, Coumar MS. Targeting survivin for cancer therapy: Strategies, small molecule inhibitors and vaccine based therapeutics in development. Life Sci 2023; 335:122260. [PMID: 37963509 DOI: 10.1016/j.lfs.2023.122260] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/16/2023]
Abstract
Survivin is a member of the family of inhibitors of apoptosis proteins (IAPs). It is involved in the normal mitotic process and acts as an anti-apoptotic molecule. While terminally differentiated normal tissues lack survivin, several human malignancies have significant protein levels. Resistance to chemotherapy and radiation in tumor cells is associated with survivin expression. Decreased tumor development, apoptosis, and increased sensitivity to chemotherapy and radiation are all effects of downregulating survivin expression or activity. As a prospective cancer treatment, small molecules targeting the transcription and translation of survivin and molecules that can directly bind with the survivin are being explored both in pre-clinical and clinics. Pre-clinical investigations have found and demonstrated the effectiveness of several small-molecule survivin inhibitors. Unfortunately, these inhibitors have also been shown to have off-target effects, which could limit their clinical utility. In addition to small molecules, several survivin peptide vaccines are currently under development. These vaccines are designed to elicit a cytotoxic T-cell response against survivin, which could lead to the destruction of tumor cells expressing survivin. Some survivin-based vaccines are advancing through Phase II clinical studies. Overall, survivin is a promising cancer drug target. However, challenges still need to be addressed before the survivin targeted therapies can be widely used in the clinics.
Collapse
Affiliation(s)
- Sree Karani Kondapuram
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India
| | - Hema Kasthuri Ramachandran
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India
| | - Hemant Arya
- Institute for Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr University Bochum, 44780 Bochum, Germany
| | - Mohane Selvaraj Coumar
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India.
| |
Collapse
|
43
|
Effendi KY, Nasrul E, Zulqarnain I, Theodorus, Amran R, Manan H, Abadi A, Usman F, Kesty C. Diagnostic Test of Transforming Growth Factor-Beta 1 (TGF- β1) in Menstrual Blood with Endometriosis. Obstet Gynecol Int 2023; 2023:9970818. [PMID: 38116460 PMCID: PMC10728358 DOI: 10.1155/2023/9970818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 12/21/2023] Open
Abstract
Background Endometriosis is a benign disorder that is generally defined as the presence of endometrial glands and stroma outside their normal location. TGF-β1 is found in stromal cells and its expression is increased in epithelial cells of endometriotic cysts. Endometriosis diagnostics take a long time, so new markers are needed to diagnose endometriosis. This study aims to determine the diagnostic value of TGF-β1 in menstrual blood in diagnosing endometriosis. Method Diagnostic tests to compare eutopic endometrial TGF-β1 levels from menstrual blood of patients with suspected endometriosis were undertaken in the Obstetrics and Gynecology Department of Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Sriwijaya University, Palembang, from July 2019 to November 2020. 50 patients who were suspected with endometriosis met the inclusion criteria. Comparison of TGF-β1 levels between endometriosis and nonendometriosis patients was analyzed using the Mann-Whitney test. The cutoff point of the TGF-β1 level towards the histopathological outcome was obtained using the ROC curve. Data analysis was performed by using SPSS version 22.0. Results In this study, endometriosis patients were 31.6 ± 6.55 years of age with a range of 20 to 46 years. In statistical analysis, there was no difference in BMI (p = 0.181) and BMI classification (p = 0.207), the history of contraception (p = 0.097), infertility (p = 1.000), and dysmenorrhoea (p = 1.000) between endometriosis and nonendometriosis patients. In the study, there were differences in TGF-β1 between endometriosis and nonendometriosis patients (p ≤ 0.001). By using the ROC curve, the cutoff point for TGF-β1 levels has the best sensitivity and specificity, which is 515 ng/ml. The TGF-β1 level has a sensitivity of 80%, a specificity of 90%, a positive predictive value (PPV) of 0.969, a negative predictive value (NPV) of 0.529, a positive likelihood ratio of 8, a negative likelihood ratio of 0.222, and an accuracy of 0.820 to the endometriosis outcome. Conclusion It can be concluded that the TGF-β1 level has a very good diagnostic value in establishing endometriosis diagnostics. This trial is registered with ISRCTN72218532.
Collapse
Affiliation(s)
- Kemas Yusuf Effendi
- Division of Reproductive, Endocrinology, and Infertility, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Excellena Nasrul
- Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Iskandar Zulqarnain
- Division of Reproductive, Endocrinology, and Infertility, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Theodorus
- Medical and Health Research Unit, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Rizani Amran
- Division of Reproductive, Endocrinology, and Infertility, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Heriyadi Manan
- Division of Reproductive, Endocrinology, and Infertility, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Adnan Abadi
- Division of Reproductive, Endocrinology, and Infertility, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Fatimah Usman
- Division of Reproductive, Endocrinology, and Infertility, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| | - Cindy Kesty
- Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang, South Sumatera, Indonesia
| |
Collapse
|
44
|
Ishitani T. Cadherin-linked morphogen gradient actualizes robust tissue patterning. Curr Opin Cell Biol 2023; 85:102275. [PMID: 37944424 DOI: 10.1016/j.ceb.2023.102275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/30/2023] [Accepted: 10/14/2023] [Indexed: 11/12/2023]
Abstract
Morphogen gradients govern tissue patterning. These gradients provide positional information, instructing cells to adopt distinct fates. Over the past few decades, extensive studies have revealed the detailed mechanisms by which morphogens generate tissue patterns. However, the communication between morphogen-receiving cells is still poorly understood. Here, I describe how cadherin-mediated cell competition ensures robust morphogen-gradient formation. In normal zebrafish embryos, unfit cells with abnormal Wnt signaling activity spontaneously appear and produce a noisy morphogen gradient. These unfit cells communicate with neighboring cells through cadherins and are subsequently killed by cell competition. This process of killing unfit cells corrects noisy gradients to support reproducible patterning. I also discuss the significance of cell-competition-mediated morphogen-gradient correction from the perspectives of evolution and disease biology.
Collapse
Affiliation(s)
- Tohru Ishitani
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
45
|
Fonseca Teixeira A, Wu S, Luwor R, Zhu HJ. A New Era of Integration between Multiomics and Spatio-Temporal Analysis for the Translation of EMT towards Clinical Applications in Cancer. Cells 2023; 12:2740. [PMID: 38067168 PMCID: PMC10706093 DOI: 10.3390/cells12232740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is crucial to metastasis by increasing cancer cell migration and invasion. At the cellular level, EMT-related morphological and functional changes are well established. At the molecular level, critical signaling pathways able to drive EMT have been described. Yet, the translation of EMT into efficient diagnostic methods and anti-metastatic therapies is still missing. This highlights a gap in our understanding of the precise mechanisms governing EMT. Here, we discuss evidence suggesting that overcoming this limitation requires the integration of multiple omics, a hitherto neglected strategy in the EMT field. More specifically, this work summarizes results that were independently obtained through epigenomics/transcriptomics while comprehensively reviewing the achievements of proteomics in cancer research. Additionally, we prospect gains to be obtained by applying spatio-temporal multiomics in the investigation of EMT-driven metastasis. Along with the development of more sensitive technologies, the integration of currently available omics, and a look at dynamic alterations that regulate EMT at the subcellular level will lead to a deeper understanding of this process. Further, considering the significance of EMT to cancer progression, this integrative strategy may enable the development of new and improved biomarkers and therapeutics capable of increasing the survival and quality of life of cancer patients.
Collapse
Affiliation(s)
- Adilson Fonseca Teixeira
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia (S.W.); (R.L.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211800, China
| | - Siqi Wu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia (S.W.); (R.L.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211800, China
| | - Rodney Luwor
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia (S.W.); (R.L.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211800, China
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
- Health, Innovation and Transformation Centre, Federation University, Ballarat, VIC 3350, Australia
| | - Hong-Jian Zhu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia (S.W.); (R.L.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211800, China
| |
Collapse
|
46
|
Liu Z, Lei Y, Shen J, Zhao G, Wang X, Wang Y, Kudo Y, Liao J, Huang Y, Yu T. Development and validation of an immune-related gene prognostic index for lung adenocarcinoma. J Thorac Dis 2023; 15:6205-6227. [PMID: 38090291 PMCID: PMC10713328 DOI: 10.21037/jtd-23-1374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/04/2023] [Indexed: 01/02/2025]
Abstract
BACKGROUND Lung cancer is the most common malignant tumor in the world, and its prognosis is still not optimistic. The aim of this study was to establish an immune-related gene (IRG) prognostic index (IRGPI) for lung adenocarcinoma (LUAD) based on IRGs, and to explore the prognosis, molecular and immune features, and response to immune checkpoint inhibitor (ICI) therapy in IRGPI-classified different subgroups of LUAD. METHODS Based on the LUAD transcriptome RNA-sequencing data in TCGA database, the differentially expressed genes (DEGs) were selected. Subsequently, DEGs were intersected with IRGs to obtain differentially expressed immune-related genes (DEIRGs). Weighted gene co-expression network analysis (WGCNA) identified hub genes in DEIRGs. Finally, univariate and multivariate Cox regression analyses were used to build an IRGPI model. Subsequently, TCGA patients were divided into high- and low-risk groups, and the survival of patients in different groups was further analyzed. Besides, we validated the molecular and immune characteristics, relationship with immune checkpoints, angiogenesis-related genes, and immune subtypes distribution in different subgroups. Meanwhile, we further validated the response to ICI therapy in different subgroups. RESULTS The IRGPI was constructed based on 13 DEIRGs. Compared with the low-risk group, overall survival (OS) was lower in the high-risk group, and the high-risk score was independently associated with poorer OS. Besides, the high-risk score was associated with cell cycle pathway, high mutation rate of TP53 and KRAS, high infiltration of M0 macrophages, and immunosuppressive state, and these patients had poorer prognosis but the TIDE score of the high-risk group was lower than that of the other group, which means that the high-risk group could benefit more from ICI treatment. In contrast, the low-risk score was related to low mutation rate of TP53 and KRAS, high infiltration of plasma cells, and immunoactive state, and these patients had better prognosis but the low-risk group less benefit from ICI treatment based on the results of TIDE score. CONCLUSIONS IRGPI is a prospective biomarker based on IRGs that can distinguish high- and low-risk groups to predict patient prognosis, help characterize the tumor immune microenvironment, and evaluate the benefit of ICI therapy in LUAD.
Collapse
Affiliation(s)
- Zitao Liu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yujie Lei
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Junting Shen
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guangqiang Zhao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xi Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yutian Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yujin Kudo
- Department of Surgery, Tokyo Medical University, Tokyo, Japan
| | - Jun Liao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunchao Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tingdong Yu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
47
|
Fernandes R, Costa C, Fernandes R, Barros AN. Inflammation in Prostate Cancer: Exploring the Promising Role of Phenolic Compounds as an Innovative Therapeutic Approach. Biomedicines 2023; 11:3140. [PMID: 38137361 PMCID: PMC10740737 DOI: 10.3390/biomedicines11123140] [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: 10/31/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Prostate cancer (PCa) remains a significant global health concern, being a major cause of cancer morbidity and mortality worldwide. Furthermore, profound understanding of the disease is needed. Prostate inflammation caused by external or genetic factors is a central player in prostate carcinogenesis. However, the mechanisms underlying inflammation-driven PCa remain poorly understood. This review dissects the diagnosis methods for PCa and the pathophysiological mechanisms underlying the disease, clarifying the dynamic interplay between inflammation and leukocytes in promoting tumour development and spread. It provides updates on recent advances in elucidating and treating prostate carcinogenesis, and opens new insights for the use of bioactive compounds in PCa. Polyphenols, with their noteworthy antioxidant and anti-inflammatory properties, along with their synergistic potential when combined with conventional treatments, offer promising prospects for innovative therapeutic strategies. Evidence from the use of polyphenols and polyphenol-based nanoparticles in PCa revealed their positive effects in controlling tumour growth, proliferation, and metastasis. By consolidating the diverse features of PCa research, this review aims to contribute to increased understanding of the disease and stimulate further research into the role of polyphenols and polyphenol-based nanoparticles in its management.
Collapse
Affiliation(s)
- Raquel Fernandes
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Cátia Costa
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Rúben Fernandes
- FP-I3ID, Instituto de Investigação, Inovação e Desenvolvimento, FP-BHS, Biomedical and Health Sciences, Universidade Fernando Pessoa, 4249-004 Porto, Portugal;
- CECLIN, Centro de Estudos Clínicos, Hospital Fernando Pessoa, 4420-096 Gondomar, Portugal
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana Novo Barros
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| |
Collapse
|
48
|
Pavičić I, Rokić F, Vugrek O. Effects of S-Adenosylhomocysteine Hydrolase Downregulation on Wnt Signaling Pathway in SW480 Cells. Int J Mol Sci 2023; 24:16102. [PMID: 38003292 PMCID: PMC10671441 DOI: 10.3390/ijms242216102] [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: 09/13/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
S-adenosylhomocysteine hydrolase (AHCY) deficiency results mainly in hypermethioninemia, developmental delay, and is potentially fatal. In order to shed new light on molecular aspects of AHCY deficiency, in particular any changes at transcriptome level, we enabled knockdown of AHCY expression in the colon cancer cell line SW480 to simulate the environment occurring in AHCY deficient individuals. The SW480 cell line is well known for elevated AHCY expression, and thereby represents a suitable model system, in particular as AHCY expression is regulated by MYC, which, on the other hand, is involved in Wnt signaling and the regulation of Wnt-related genes, such as the β-catenin co-transcription factor LEF1 (lymphoid enhancer-binding factor 1). We selected LEF1 as a potential target to investigate its association with S-adenosylhomocysteine hydrolase deficiency. This decision was prompted by our analysis of RNA-Seq data, which revealed significant changes in the expression of genes related to the Wnt signaling pathway and genes involved in processes responsible for epithelial-mesenchymal transition (EMT) and cell proliferation. Notably, LEF1 emerged as a common factor in these processes, showing increased expression both on mRNA and protein levels. Additionally, we show alterations in interconnected signaling pathways linked to LEF1, causing gene expression changes with broad effects on cell cycle regulation, tumor microenvironment, and implications to cell invasion and metastasis. In summary, we provide a new link between AHCY deficiency and LEF1 serving as a mediator of changes to the Wnt signaling pathway, thereby indicating potential connections of AHCY expression and cancer cell phenotype, as Wnt signaling is frequently associated with cancer development, including colorectal cancer (CRC).
Collapse
Affiliation(s)
| | | | - Oliver Vugrek
- Laboratory for Advanced Genomics, Divison of Molecular Medicine, Institute Ruđer Bošković, Bijenička Cesta 54, 10000 Zagreb, Croatia; (I.P.); (F.R.)
| |
Collapse
|
49
|
Ramzan A, Yousaf MA, Rashid MU, Basheera S, Malkani N. In-silico prediction of TGF-β1 non-synonymous variants and their impact on binding affinity to Fresolimumab. J Biomol Struct Dyn 2023; 42:12214-12227. [PMID: 37817532 DOI: 10.1080/07391102.2023.2268198] [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: 05/02/2023] [Accepted: 10/03/2023] [Indexed: 10/12/2023]
Abstract
TGF-β1 is a potent immunoregulatory cytokine that plays diverse roles in development, bone healing, fibrosis, and cancer. However, characterizing TGF-β1 gene variants is challenging because the structural and functional consequences of these variants are still undetermined. In this study, we aimed to perform an in-silico analysis of TGF-β1 non-synonymous variants and their pathogenic effects on the TGF-β1 protein. A total of 10,252 TGF-β1 SNPs were collected from the NCBI dbSNP database and in-silico tools (SIFT, PROVEAN, Mutation Taster, ClinVar, PolyPhen-2, CScape, MutPred, and ConSurf) were used. The in-silico predicted potential variants were further investigated for their binding to the TGF-β1 targeting drug "Fresolimumab". Molecular docking was performed using HADDOCK and confirmed by PRODIGY and PDBsum. The in-silico analysis predicted four potential TGF-β1 nsSNPs: E47G in the LAP domain of the propeptide and I22T, L28F, and E35D in the mature TGF-β1 peptide. HADDOCK and molecular dynamics simulations revealed that the I22T and E35D variants have higher binding affinity for Fresolimumab as compared to the wild type and L28F variants. Molecular dynamics simulations (100 ns) and principal component analysis showed that TGF-β1 variants influenced the protein structure and caused variations in the internal dynamics of protein complexes with the antibody. Among them, the E35D variant significantly destabilized the TGF-β1 protein structure, resulting in rearrangement in the binding site and affecting the interactions with the Fresolimumab. This study identified four variants that can affect the TGF-β1 protein structure and result in functional consequences such as impaired response to Fresolimumab.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ammara Ramzan
- Department of Zoology, GC University, Lahore, Pakistan
| | - Muhammad Abrar Yousaf
- Section of Biology and Genetics, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Muhammad Usman Rashid
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH & RC), Lahore, Pakistan
| | - Shefin Basheera
- Department of Biotechnology and Bioinformatics, Thiruvananthapuram, India
| | - Naila Malkani
- Department of Zoology, GC University, Lahore, Pakistan
| |
Collapse
|
50
|
Kobayashi K, Takemura RD, Miyamae J, Mitsui I, Murakami K, Kutara K, Saeki K, Kanda T, Okamura Y, Sugiyama A. Phenotypic and molecular characterization of novel pulmonary adenocarcinoma cell lines established from a dog. Sci Rep 2023; 13:16823. [PMID: 37798461 PMCID: PMC10556002 DOI: 10.1038/s41598-023-44062-1] [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: 06/21/2023] [Accepted: 10/03/2023] [Indexed: 10/07/2023] Open
Abstract
Canine pulmonary adenocarcinoma (PAC) resembles human lung tumors in never-smokers, but it is rarer than human pulmonary adenocarcinoma. Therefore, research on canine PAC is challenging. In the present study, we successfully established various novel canine PAC cell lines from a single lesion in a dog, including two parent cell lines and fourteen cloned cell lines, and characterized their cellular properties in vitro. Several of these cell lines showed epithelial-mesenchymal transition (EMT)-like and/or cancer stem cell (CSCs)-like phenotypes. We additionally assessed the sensitivity of the cells to vinorelbine in vitro. Three clonal lines, two of which showed EMT- and CSC-like phenotypes, were resistant to vinorelbine. Furthermore, we evaluated the expression and activation status of EGFR, HER2, and Ras signaling factors. The findings indicated that the cell lines we established preserved the expression and activation of these factors to varying extents. These novel canine PAC cell lines can be utilized in future research for understanding the pathogenesis and development of treatments for canine PAC.
Collapse
Affiliation(s)
- Kosuke Kobayashi
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan.
| | - Reika Deja Takemura
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Jiro Miyamae
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Ikki Mitsui
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Kohei Murakami
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Kenji Kutara
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Kohei Saeki
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Teppei Kanda
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Yasuhiko Okamura
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| | - Akihiko Sugiyama
- Faculty of Veterinary Medicine, Okayama University of Science, Ikoino-oka 1-3, Imabari Ehime, Japan
| |
Collapse
|