1
|
Qiu LX, Yu Q, Zhou HQ, Fan WH, Zheng JJ, Yang YL, Zhang WZ, Cao X, Yang H. TMT-based quantitative proteomics reveals the genetic mechanisms of secondary hair follicle development in fine-wool sheep. PLoS One 2025; 20:e0315637. [PMID: 39913466 PMCID: PMC11801579 DOI: 10.1371/journal.pone.0315637] [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: 07/24/2024] [Accepted: 11/29/2024] [Indexed: 02/09/2025] Open
Abstract
The development of secondary hair follicles influences the quality of sheep wool. However, the mechanism by which proteins mediate the fetal development of secondary hair follicles remains unknown. In this study, the histomorphology of secondary hair follicles was analyzed over four stages of fetal development (75, 85, 95, and 105 gestational days). TMT-based quantitative proteomics was used to compare the differential protein profiles of skin tissues between consecutive developmental periods (75 versus 85, 85 versus 95, and 95 versus 105 gestational days). We found that the density of secondary hair follicles and the secondary hair follicles/primary hair follicles ratio increased from 85 to 105 gestational days. Bioinformatic analysis identified 238, 35, and 348 differentially expressed proteins in the respective comparison periods. Focal adhesion, ECM-receptor interaction, and the estrogen signaling pathway all played important roles in secondary hair follicle development. COL1A1, THBS3, ITGA6, COL6A1, and THBS4 were identified as potential candidate proteins in the initiation of secondary hair follicles. This study provides valuable proteomics data on secondary hair follicle development and thus has deepened our understanding of the molecular mechanisms underlying wool quality traits in fine-wool sheep.
Collapse
Affiliation(s)
- Li-Xia Qiu
- College of Animal Science and Technology, Northwest Minzu University, Lanzhou, Gansu, China
| | - Qian Yu
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, Xinjiang, China
| | - Hua-Qian Zhou
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, Xinjiang, China
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Wen-Hua Fan
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, Xinjiang, China
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Jing-Jing Zheng
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, Xinjiang, China
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Yong-Lin Yang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, Xinjiang, China
| | - Wen-Zhe Zhang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, Xinjiang, China
| | - Xin Cao
- College of Animal Science and Technology, Northwest Minzu University, Lanzhou, Gansu, China
| | - Hua Yang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, Xinjiang, China
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
2
|
Xiao X, Long F, Yu S, Wu W, Nie D, Ren X, Li W, Wang X, Yu L, Wang P, Wang G. Col1A1 as a new decoder of clinical features and immune microenvironment in ovarian cancer. Front Immunol 2025; 15:1496090. [PMID: 39845977 PMCID: PMC11750837 DOI: 10.3389/fimmu.2024.1496090] [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: 09/16/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025] Open
Abstract
Backgrounds Collagen type I alpha 1 chain (COL1A1) is a key protein encoding fibrillar collagen, playing a crucial role in the tumor microenvironment (TME) due to its complex functions and close association with tumor invasiveness. This has made COL1A1 a focal point in cancer biology research. However, studies investigating the relationship between COL1A1 expression levels and clinical characteristics of ovarian cancer (OC) remain limited. Methods This study integrated resources from publicly available online databases and immunohistochemistry (IHC) techniques to analyze and validate COL1A1 expression in OC tissues, and evaluated its potential association with clinical features in OC patients. The prognostic value of COL1A1 was assessed using Kaplan-Meier (KM) survival curve analysis. The TIMER and TISIDB databases to explore the potential relationship between COL1A1 expression and immune microenvironment in OC tissues. The LinkedOmics and INPUT2 databases were used to analyze differential gene expression in OC, This was followed by enrichment analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) annotations to identify and predict potential signaling pathways associated with COL1A1. Results Our study demonstrated that COL1A1 expression was significantly elevated in OC tissues compared to normal ovarian tissues. This elevated expression was closely associated with tumor metastasis, poor prognosis, and advanced pathological stages in OC patients. Moreover, COL1A1 expression showed a significant correlation with immune cell infiltration and the expression of immune-related genes within the TME.Further analyses revealed that COL1A1 and its co-expressed genes were primarily enriched in key signaling pathways involved in OC invasion, metastasis, and angiogenesis, indicating its potential role in driving OC progression. Conclusions Our study found that upregulation of COL1A1 expression is significantly associated with lymph node metastasis of OC and can affect the immune microenvironment. Based on this, COL1A1 could serve as a promising biomarker for OC prognosis and provide a new perspective for the development of potential immunotherapies for patients with OC.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Gynecology, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Shaolan Yu
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Wengjuan Wu
- Department of Gynecology, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Dayan Nie
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaoyan Ren
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Wen Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xujuan Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Ling Yu
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Pinghan Wang
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Gang Wang
- Department of Gynecology, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Roy NS, Kumari M, Alam K, Bhattacharya A, Kaity S, Kaur K, Ravichandiran V, Roy S. Development of bioengineered 3D patient derived breast cancer organoid model focusing dynamic fibroblast-stem cell reciprocity. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 7:012007. [PMID: 39662055 DOI: 10.1088/2516-1091/ad9dcb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024]
Abstract
Three-dimensional (3D) models, such as tumor spheroids and organoids, are increasingly developed by integrating tissue engineering, regenerative medicine, and personalized therapy strategies. These advanced 3Din-vitromodels are not merely endpoint-driven but also offer the flexibility to be customized or modulated according to specific disease parameters. Unlike traditional 2D monolayer cultures, which inadequately capture the complexities of solid tumors, 3D co-culture systems provide a more accurate representation of the tumor microenvironment. This includes critical interactions with mesenchymal stem/stromal cells (MSCs) and induced pluripotent stem cells (iPSCs), which significantly modulate cancer cell behavior and therapeutic responses. Most of the findings from the co-culture of Michigan Cancer Foundation-7 breast cancer cells and MSC showed the formation of monolayers. Although changes in the plasticity of MSCs and iPSCs caused by other cells and extracellular matrix (ECM) have been extensively researched, the effect of MSCs on cancer stem cell (CSC) aggressiveness is still controversial and contradictory among different research communities. Some researchers have argued that CSCs proliferate more, while others have proposed that cancer spread occurs through dormancy. This highlights the need for further investigation into how these interactions shape cancer aggressiveness. The objective of this review is to explore changes in cancer cell behavior within a 3D microenvironment enriched with MSCs, iPSCs, and ECM components. By describing various MSC and iPSC-derived 3D breast cancer models that replicate tumor biology, we aim to elucidate potential therapeutic targets for breast cancer. A particular focus of this review is the Transwell system, which facilitates understanding how MSCs and iPSCs affect critical processes such as migration, invasion, and angiogenesis. The gradient formed between the two chambers is based on diffusion, as seen in the human body. Once optimized, this Transwell model can serve as a high-throughput screening platform for evaluating various anticancer agents. In the future, primary cell-based and patient-derived 3D organoid models hold promise for advancing personalized medicine and accelerating drug development processes.
Collapse
Affiliation(s)
- Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Mamta Kumari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Department of Anatomy & Regenerative Medicine, Tissue Engineering Research Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| |
Collapse
|
4
|
Mu Y, Lian C, Chen X, Yang X, Li D, Zhang Y. Cutaneous squamous cell carcinoma-derived exosomal MicroRNA-31 acts as an oncogene by targeting the tumor suppressor RhoBTB1. Arch Dermatol Res 2024; 317:114. [PMID: 39673615 DOI: 10.1007/s00403-024-03558-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 09/29/2024] [Accepted: 11/21/2024] [Indexed: 12/16/2024]
Abstract
Tumor-derived exosomes that transport MicroRNAs significantly influence cutaneous squamous cell carcinoma(CSCC) progression. However, the molecular mechanisms of exosomal MicroRNA-31 regulation of CSCC are mostly undefined. To determine whether a targeting relationship exists between MicroRNA-31 (miR-31) in CSCC-derived exosomes and the tumor suppressor RhoBTB1, and the regulatory effect of the relationship on tumor cells. Immunoblotting and quantitative PCR were used to measure miR-31 and RhoBTB1 levels in various cells and exosomes. Differential ultracentrifugation was used to isolate exosomes. MTT and Transwell assays were used to assess cell proliferation, migration, and invasion. Dual luciferase reporter assays were used to assess the direct interaction between miR-31 and the tumor suppressor RhoBTB1 in cutaneous squamous cell carcinoma (CSCC)-derived exosomes. Compared with a human skin keratinocyte cell line, in CSCC cell lines RhoBTB1 was downregulated and miR-31 levels were elevated. Exosomal miR-31 from CSCC cell lines directly targeted RhoBTB1 by binding to the 3' UTR of RhoBTB1. This interaction suppressed expression of RhoBTB1 and enhanced CSCC cell proliferation, migration, and invasion. MicroRNA-31 in CSCC-derived exosomes can enhance CSCC cell proliferation, migration, and invasion by suppressing expression of RhoBTB1. This finding explains, in part, the molecular mechanism of CSCC. Investigative approaches focused on suppressing miR-31 or enhancing RhoBTB1 signaling pathways are promising avenues for developing targeted therapies for CSCC.
Collapse
Affiliation(s)
- Yanan Mu
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia Medical University, Xinhua Street, Hohhot, 010030, China
| | - Chen Lian
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia Medical University, Xinhua Street, Hohhot, 010030, China
| | - Xinghui Chen
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia Medical University, Xinhua Street, Hohhot, 010030, China
| | - Xueying Yang
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia Medical University, Xinhua Street, Hohhot, 010030, China
| | - Dongxia Li
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia Medical University, Xinhua Street, Hohhot, 010030, China.
| | - Yi Zhang
- Department of Dermatology, The Affiliated Hospital of Inner Mongolia Medical University, Xinhua Street, Hohhot, 010030, China.
| |
Collapse
|
5
|
Shi J, Lin Z, Zheng Z, Chen M, Huang X, Wang J, Li M, Shao J. Glutamine metabolism promotes human trophoblast cell invasion via COL1A1 mediated by PI3K-AKT pathway. J Reprod Immunol 2024; 166:104321. [PMID: 39243705 DOI: 10.1016/j.jri.2024.104321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/13/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024]
Abstract
Abnormal trophoblast invasion function is an important cause of recurrent spontaneous abortion (RSA). Recent research has revealed a connection between glutamine metabolism and RSA. However, the interplay between these three factors and their related mechanisms remains unclear. To address this issue, we collected villus tissues from 10 healthy women with induced abortion and from 10 women with RSA to detect glutamine metabolism. Then, the trophoblast cell line HTR-8/SVneo was used in vitro to explore the effect of glutamine metabolism on trophoblast cells invasion, which was tested by transwell assay. We found that the concentration of glutamine in the villi of the normal pregnancy group was significantly higher than that in the RSA group. Correspondingly, the expression levels of key enzymes involved in glutamine synthesis and catabolism, including glutamine synthetase and glutaminase, were significantly higher in the villi of the normal pregnancy group. Regarding trophoblast cells, glutamine markedly enhanced the proliferative and invasive abilities of HTR-8/SVneo cells. Additionally, collagen type I alpha 1 (COL1A1) was confirmed to be a downstream target of glutamine, and glutamine also activated the PI3K-AKT pathway in HTR-8/SVneo cells. These findings indicate that glutamine metabolism facilitates the invasion of trophoblasts by up-regulating COL1A1 expression through the activation of the PI3K-AKT pathway, but the specific mechanism of COL1A1 requires further study.
Collapse
Affiliation(s)
- Jialu Shi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200090, China
| | - Zhi Lin
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200090, China
| | - Zimeng Zheng
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Min Chen
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310000, China
| | - Xu Huang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Jiarui Wang
- Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China.
| | - Jun Shao
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200090, China.
| |
Collapse
|
6
|
Kim K, Kim DG, Kim YJ. RhoBTB3 Functions as a Novel Regulator of Autophagy by Suppressing AMBRA1 Stability. Cells 2024; 13:1659. [PMID: 39404422 PMCID: PMC11475653 DOI: 10.3390/cells13191659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Autophagy is essential for cell survival and cellular homeostasis under various stress conditions. Therefore, autophagy dysfunction is associated with the pathogenesis of various human diseases. We explored the regulatory role of RhoBTB3 in autophagy and its interaction with activating molecules in AMBRA1. RhoBTB3 deficiency was found to induce autophagy, while its overexpression inhibited autophagy induction. Through immunoprecipitation and mass spectrometry, AMBRA1 was identified as a substrate of RhoBTB3. The study revealed that RhoBTB3 regulates AMBRA1 stability by influencing its protein levels without affecting its mRNA levels. RhoBTB3 induced the ubiquitination of AMBRA1, leading to proteasome-mediated degradation, with the ubiquitination occurring at K45 on AMBRA1 through a K27-linked ubiquitin chain. The knockdown of AMBRA1 blocked RhoBTB3 knockdown-induced autophagy, indicating the dependency of autophagy on AMBRA1. Thus, RhoBTB3 negatively regulates autophagy by mediating AMBRA1 ubiquitination and degradation, suggesting RhoBTB3 as a potential therapeutic target for autophagy-related diseases.
Collapse
Affiliation(s)
| | | | - Youn-Jae Kim
- Targeted Therapy Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
7
|
Pantazi V, Miklós V, Smith P, Oláh-Németh O, Pankotai-Bodó G, Teja Dondapati D, Ayaydin F, D'Angiolella V, Pankotai T. Prognostic potential of CUL3 ligase with differential roles in luminal A and basal type breast cancer tumors. Sci Rep 2024; 14:14912. [PMID: 38942922 PMCID: PMC11213933 DOI: 10.1038/s41598-024-65692-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024] Open
Abstract
Breast cancer is a prevalent and significant cause of mortality in women, and manifests as six molecular subtypes. Its further histologic classification into non-invasive ductal or lobular carcinoma (DCIS) and invasive carcinoma (ILC or IDC) underscores its heterogeneity. The ubiquitin-proteasome system plays a crucial role in breast cancer, with inhibitors targeting the 26S proteasome showing promise in clinical treatment. The Cullin-RING ubiquitin ligases, including CUL3, have direct links to breast cancer. This study focuses on CUL3 as a potential biomarker, leveraging high-throughput sequencing, gene expression profiling, experimental and data analysis tools. Through comprehensive analysis using databases like GEPIA2 and UALCAN, as well as TCGA datasets, CUL3's expression and its association with prognostic values were assessed. Additionally, the impact of CUL3 overexpression was explored in MCF-7 and MDA-MB-231 breast cancer cell lines, revealing distinct differences in molecular and phenotypic characteristics. We further profiled its expression and localization in breast cancer tissues identifying prominent differences between luminal A and TNBC tumors. Conclusively, CUL3 was found to be associated with cell cycle progression, and DNA damage response, exhibiting diverse roles depending on the tumor's molecular type. It exhibits a tendency to act as an oncogene in triple-negative tumors and as a tumor suppressor in luminal A types, suggesting a potential significance in breast cancer progression and therapeutic directions.
Collapse
Affiliation(s)
- Vasiliki Pantazi
- Genome Integrity and DNA Repair Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Szeged, Hungary
| | - Vanda Miklós
- Genome Integrity and DNA Repair Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Paul Smith
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Orsolya Oláh-Németh
- Genome Integrity and DNA Repair Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gabriella Pankotai-Bodó
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Divya Teja Dondapati
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Functional Cell Biology and Immunology Advanced Core Facility, University of Szeged, Szeged, Hungary
| | - Ferhan Ayaydin
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Functional Cell Biology and Immunology Advanced Core Facility, University of Szeged, Szeged, Hungary
| | | | - Tibor Pankotai
- Genome Integrity and DNA Repair Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary.
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
- Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Szeged, Hungary.
| |
Collapse
|
8
|
Lee DK, Oh J, Park HW, Gee HY. Anchorage Dependence and Cancer Metastasis. J Korean Med Sci 2024; 39:e156. [PMID: 38769921 PMCID: PMC11106561 DOI: 10.3346/jkms.2024.39.e156] [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: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
The process of cancer metastasis is dependent on the cancer cells' capacity to detach from the primary tumor, endure in a suspended state, and establish colonies in other locations. Anchorage dependence, which refers to the cells' reliance on attachment to the extracellular matrix (ECM), is a critical determinant of cellular shape, dynamics, behavior, and, ultimately, cell fate in nonmalignant and cancer cells. Anchorage-independent growth is a characteristic feature of cells resistant to anoikis, a programmed cell death process triggered by detachment from the ECM. This ability to grow and survive without attachment to a substrate is a crucial stage in the progression of metastasis. The recently discovered phenomenon named "adherent-to-suspension transition (AST)" alters the requirement for anchoring and enhances survival in a suspended state. AST is controlled by four transcription factors (IKAROS family zinc finger 1, nuclear factor erythroid 2, BTG anti-proliferation factor 2, and interferon regulatory factor 8) and can detach cells without undergoing the typical epithelial-mesenchymal transition. Notably, AST factors are highly expressed in circulating tumor cells compared to their attached counterparts, indicating their crucial role in the spread of cancer. Crucially, the suppression of AST substantially reduces metastasis while sparing primary tumors. These findings open up possibilities for developing targeted therapies that inhibit metastasis and emphasize the importance of AST, leading to a fundamental change in our comprehension of how cancer spreads.
Collapse
Affiliation(s)
- Dong Ki Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
| | - Jongwook Oh
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul, Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Li X, Jin Y, Xue J. Unveiling Collagen's Role in Breast Cancer: Insights into Expression Patterns, Functions and Clinical Implications. Int J Gen Med 2024; 17:1773-1787. [PMID: 38711825 PMCID: PMC11073151 DOI: 10.2147/ijgm.s463649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/21/2024] [Indexed: 05/08/2024] Open
Abstract
Collagen, the predominant protein constituent of the mammalian extracellular matrix (ECM), comprises a diverse family of 28 members (I-XXVIII). Beyond its structural significance, collagen is implicated in various diseases or cancers, notably breast cancer, where it influences crucial cellular processes including proliferation, metastasis, apoptosis, and drug resistance, intricately shaping cancer progression and prognosis. In breast cancer, distinct collagens exhibit differential expression profiles, with some showing heightened or diminished levels in cancerous tissues or cells compared to normal counterparts, suggesting specific and pivotal biological functions. In this review, we meticulously analyze the expression of individual collagen members in breast cancer, utilizing Transcripts Per Million (TPM) data sourced from the GEPIA2 database. Through this analysis, we identify collagens that deviate from normal expression patterns in breast cancer, providing a comprehensive overview of their expression dynamics, functional roles, and underlying mechanisms. Our findings shed light on recent advancements in understanding the intricate interplay between these aberrantly expressed collagens and breast cancer. This exploration aims to offer valuable insights for the identification of potential biomarkers and therapeutic targets, thereby advancing the prospects of more effective interventions in breast cancer treatment.
Collapse
Affiliation(s)
- Xia Li
- Department of Molecular Diagnosis, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Yue Jin
- Department of Molecular Diagnosis, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Jian Xue
- Department of Emergency Medicine, Yizheng People’s Hospital, Yangzhou, People’s Republic of China
| |
Collapse
|
10
|
Ding D, Xu C, Zhang J, Zhang Y, Xue L, Song J, Luo Z, Hong X, Wang J, Liang W, Xue X. Revealing underlying regulatory mechanisms of LINC00313 in Osimertinib-resistant LUAD cells by ceRNA network analysis. Transl Oncol 2024; 43:101895. [PMID: 38377935 PMCID: PMC10884499 DOI: 10.1016/j.tranon.2024.101895] [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: 10/03/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Osimertinib, a third-generation epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI), is the preferred treatment for EGFR-mutated lung cancer. However, acquired resistance inevitably develops. While non-coding RNAs have been implicated in lung cancer through various functions, the molecular mechanisms responsible for osimertinib resistance remain incompletely elucidated. METHODS RNA-sequencing technology was employed to determine differentially expressed lncRNAs (DE-lncRNAs) and mRNAs (DE-mRNAs) between H1975 and H1975OR cell lines. Starbase 2.0 was utilized to predict DE-lncRNA and DE-mRNA interactions, constructing ceRNA networks. Subsequently, functional and pathway enrichment analysis were performed on target DE-mRNAs to identify pathways associated with osimertinib resistance. Key target DE-mRNAs were then selected as potential risk signatures for lung adenocarcinoma (LUAD) prognostic modeling using multivariate Cox regression analyses. The Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) and immunohistochemistry staining were used for result validation. RESULTS Functional analysis revealed that the identified DE-mRNAs primarily enriched in EGFR-TKI resistance pathways, especially in the PI3K/Akt signaling pathway, where their concerted actions may lead to osimertinib resistance. Specifically, upregulation of LINC00313 enhanced COL1A1 expression by acting as a miR-218-5p sponge, triggering an upstream response that activates the PI3K/Akt pathway, potentially contributing to osimertinib resistance. Furthermore, the expressions of LINC00313 and COL1A1 were validated by qRT-PCR, and the activation of the PI3K/Akt pathway was confirmed by immunohistochemistry staining. CONCLUSIONS Our results suggest that the LINC00313/miR-218-5p/COL1A1 axis potentially contributes to osimertinib resistance through the PI3K/Akt signaling pathway, providing novel insights into the molecular mechanisms underlying acquired osimertinib resistance in LUAD. Additionally, our study may aid in the identification of potential therapeutic targets for overcoming resistance to osimertinib.
Collapse
Affiliation(s)
- Dandan Ding
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China; First People's Hospital of Foshan, Foshan, 528000, China
| | - Chenguang Xu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510182, China; Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou, 510182, China
| | - Jufeng Zhang
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Ying Zhang
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Lipeng Xue
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Jingjing Song
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510182, China
| | - Zhiming Luo
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiaoyu Hong
- Nanjing Geneseeq Technology Inc, Nanjing, 211899, China
| | - Jian Wang
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.
| | - Weicheng Liang
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Xingyang Xue
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.
| |
Collapse
|
11
|
Yu TY, Zhang G, Chai XX, Ren L, Yin DC, Zhang CY. Recent progress on the effect of extracellular matrix on occurrence and progression of breast cancer. Life Sci 2023; 332:122084. [PMID: 37716504 DOI: 10.1016/j.lfs.2023.122084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Breast cancer (BC) metastasis is an enormous challenge targeting BC therapy. The extracellular matrix (ECM), the principal component of the BC metastasis niche, is the pivotal driver of breast tumor development, whose biochemical and biophysical characteristics have attracted widespread attention. Here, we review the biological effects of ECM constituents and the influence of ECM stiffness on BC metastasis and drug resistance. We provide an overview of the relative signal transduction mechanisms, existing metastasis models, and targeted drug strategies centered around ECM stiffness. It will shed light on exploring more underlying targets and developing specific drugs aimed at ECM utilizing biomimetic platforms, which are promising for breast cancer treatment.
Collapse
Affiliation(s)
- Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo 315103, Zhejiang, PR China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| |
Collapse
|
12
|
Dumitru AV, Stoica EE, Covache-Busuioc RA, Bratu BG, Cirstoiu MM. Unraveling the Intricate Link: Deciphering the Role of the Golgi Apparatus in Breast Cancer Progression. Int J Mol Sci 2023; 24:14073. [PMID: 37762375 PMCID: PMC10531533 DOI: 10.3390/ijms241814073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer represents a paramount global health challenge, warranting intensified exploration of the molecular underpinnings influencing its progression to facilitate the development of precise diagnostic instruments and customized therapeutic regimens. Historically, the Golgi apparatus has been acknowledged for its primary role in protein sorting and trafficking within cellular contexts. However, recent findings suggest a potential link between modifications in Golgi apparatus function and organization and the pathogenesis of breast cancer. This review delivers an exhaustive analysis of this correlation. Specifically, we examine the consequences of disrupted protein glycosylation, compromised protein transport, and inappropriate oncoprotein processing on breast cancer cell dynamics. Furthermore, we delve into the impacts of Golgi-mediated secretory routes on the release of pro-tumorigenic factors during the course of breast cancer evolution. Elucidating the nuanced interplay between the Golgi apparatus and breast cancer can pave the way for innovative therapeutic interventions and the discovery of biomarkers, potentially enhancing the diagnostic, prognostic, and therapeutic paradigms for afflicted patients. The advancement of such research could substantially expedite the realization of these objectives.
Collapse
Affiliation(s)
- Adrian Vasile Dumitru
- Department of Pathology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| | - Evelina-Elena Stoica
- Department of Obstetrics and Gynaecology, University Emergency Hospital, 050098 Bucharest, Romania;
| | | | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Monica-Mihaela Cirstoiu
- Department of Obstetrics and Gynaecology, University Emergency Hospital, 050098 Bucharest, Romania;
- Department of Obstetrics and Gynaecology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
13
|
Ahn S, Kwon A, Oh Y, Rhee S, Song WK. Microtubule Acetylation-Specific Inhibitors Induce Cell Death and Mitotic Arrest via JNK/AP-1 Activation in Triple-Negative Breast Cancer Cells. Mol Cells 2023; 46:387-398. [PMID: 36794420 PMCID: PMC10258459 DOI: 10.14348/molcells.2023.2192] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/17/2023] Open
Abstract
Microtubule acetylation has been proposed as a marker of highly heterogeneous and aggressive triple-negative breast cancer (TNBC). The novel microtubule acetylation inhibitors GM-90257 and GM-90631 (GM compounds) cause TNBC cancer cell death but the underlying mechanisms are currently unknown. In this study, we demonstrated that GM compounds function as anti-TNBC agents through activation of the JNK/AP-1 pathway. RNA-seq and biochemical analyses of GM compound-treated cells revealed that c-Jun N-terminal kinase (JNK) and members of its downstream signaling pathway are potential targets for GM compounds. Mechanistically, JNK activation by GM compounds induced an increase in c-Jun phosphorylation and c-Fos protein levels, thereby activating the activator protein-1 (AP-1) transcription factor. Notably, direct suppression of JNK with a pharmacological inhibitor alleviated Bcl2 reduction and cell death caused by GM compounds. TNBC cell death and mitotic arrest were induced by GM compounds through AP-1 activation in vitro. These results were reproduced in vivo, validating the significance of microtubule acetylation/JNK/AP-1 axis activation in the anti-cancer activity of GM compounds. Moreover, GM compounds significantly attenuated tumor growth, metastasis, and cancer-related death in mice, demonstrating strong potential as therapeutic agents for TNBC.
Collapse
Affiliation(s)
- Suyeon Ahn
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Ahreum Kwon
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Youngsoo Oh
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Woo Keun Song
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| |
Collapse
|
14
|
Shikhevich S, Chadaeva I, Khandaev B, Kozhemyakina R, Zolotareva K, Kazachek A, Oshchepkov D, Bogomolov A, Klimova NV, Ivanisenko VA, Demenkov P, Mustafin Z, Markel A, Savinkova L, Kolchanov NA, Kozlov V, Ponomarenko M. Differentially Expressed Genes and Molecular Susceptibility to Human Age-Related Diseases. Int J Mol Sci 2023; 24:ijms24043996. [PMID: 36835409 PMCID: PMC9966505 DOI: 10.3390/ijms24043996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Mainstream transcriptome profiling of susceptibility versus resistance to age-related diseases (ARDs) is focused on differentially expressed genes (DEGs) specific to gender, age, and pathogeneses. This approach fits in well with predictive, preventive, personalized, participatory medicine and helps understand how, why, when, and what ARDs one can develop depending on their genetic background. Within this mainstream paradigm, we wanted to find out whether the known ARD-linked DEGs available in PubMed can reveal a molecular marker that will serve the purpose in anyone's any tissue at any time. We sequenced the periaqueductal gray (PAG) transcriptome of tame versus aggressive rats, identified rat-behavior-related DEGs, and compared them with their known homologous animal ARD-linked DEGs. This analysis yielded statistically significant correlations between behavior-related and ARD-susceptibility-related fold changes (log2 values) in the expression of these DEG homologs. We found principal components, PC1 and PC2, corresponding to the half-sum and the half-difference of these log2 values, respectively. With the DEGs linked to ARD susceptibility and ARD resistance in humans used as controls, we verified these principal components. This yielded only one statistically significant common molecular marker for ARDs: an excess of Fcγ receptor IIb suppressing immune cell hyperactivation.
Collapse
Affiliation(s)
- Svetlana Shikhevich
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Irina Chadaeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Bato Khandaev
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Rimma Kozhemyakina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Karina Zolotareva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Anna Kazachek
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Dmitry Oshchepkov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Anton Bogomolov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Natalya V. Klimova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Vladimir A. Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Pavel Demenkov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Zakhar Mustafin
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Arcady Markel
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Ludmila Savinkova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Vladimir Kozlov
- Research Institute of Fundamental and Clinical Immunology (RIFCI) SB RAS, Novosibirsk 630099, Russia
| | - Mikhail Ponomarenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- Correspondence: ; Tel.: +7-(383)-363-4963 (ext. 1311)
| |
Collapse
|
15
|
Xiang G, Huang L, Zhang X, Wang N, Wang H, Mu Y, Li K, Liu Z. Molecular Characteristics and Promoter Analysis of Porcine COL1A1. Genes (Basel) 2022; 13:1971. [PMID: 36360208 PMCID: PMC9689670 DOI: 10.3390/genes13111971] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 03/25/2024] Open
Abstract
COL1A1 encodes the type I collagen α1 chain, which shows the highest abundance among members of the collagen family and is widely expressed in different mammalian cells and tissues. However, its molecular characteristics are not completely elucidated. In this study, the molecular profiles of COL1A1 and characteristics of the COL1A1 protein were investigated using a promoter activity assay and multiple bioinformatics tools. The results showed that the 5' flanking region of porcine COL1A1 contained two CpG islands, five core promoter sequences, and twenty-six transcription factor-binding sites. In the luciferase assay, the upstream 294 bp region of the initiation codon of COL1A1 showed the highest activity, confirming that this section is the core region of the porcine COL1A1 promoter. Bioinformatic analysis revealed that COL1A1 is a negatively charged, hydrophilic secreted protein. It does not contain a transmembrane domain and is highly conserved in humans, mice, sheep, and pigs. Protein interaction analysis demonstrated that the interaction coefficient of COL1A1 with COL1A2, COL3A1, ITGB1, and ITGA2 was greater than 0.9, suggesting that this protein plays a crucial role in collagen structure formation and cell adhesion. These results provide a theoretical basis for further investigation of the functions of porcine COL1A1.
Collapse
Affiliation(s)
- Guangming Xiang
- Key Laboratory of Animal Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lei Huang
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Xiuling Zhang
- Key Laboratory of Animal Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Wang
- Key Laboratory of Animal Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Hui Wang
- Key Laboratory of Animal Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yulian Mu
- Key Laboratory of Animal Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kui Li
- Key Laboratory of Animal Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Zhiguo Liu
- Key Laboratory of Animal Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|