1
|
Wang Y, Wang Z, Pan J, Wang H, Lei Z, Liu J, Zou J, Lv H, Luan F. Reconstruction of the lncRNA-miRNA-mRNA network based on competitive endogenous RNA reveals functional miRNAs and lncRNAs in burns and keloids. PLoS One 2025; 20:e0320855. [PMID: 40203071 PMCID: PMC11981201 DOI: 10.1371/journal.pone.0320855] [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: 12/10/2024] [Accepted: 02/14/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUNDS Long non-coding RNAs (lncRNAs) exert their pharmacological functions by serving as sponges for related microRNAs (miRNAs), thereby modulating gene expression. Nevertheless, the regulatory roles of the lncRNA-mediated competing endogenous RNA (ceRNA) mechanism in the interplay between burns and keloids remain largely elusive. OBJECTIVE To construct the ceRNA regulatory network of burns, leveraging network pharmacology and bioinformatics analyses. RESULTS 3576 DELs (Differentially Expressed lncRNAs), 1427 DEMis (Differentially Expressed miRNAs), and 2555 DEMs (Differentially Expressed mRNAs) were identified as differentially expressed genes. A ceRNA network composed of DELs-DEMis-DEMs in burns and keloids was constructed, with a prominent sub-network consisting of 23 DELs, 330 DEMs, and 8 DEMis. Subsequently, the clusterProfiler package in the R programming language was utilized to perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The sub-network within the ceRNA network was extracted, in which three lncRNAs, namely lnc-WRB, lnc-SCNN1G, and LINC00271, and three miRNAs, namely hsa-miR-21, hsa-miR-34a, and hsa-miR-155, were identified as key genes. CONCLUSION All nodes within the sub-ceRNA network exert either a direct or an indirect influence on the pathological processes of burns and post-burn keloids. The current study successfully constructed the ceRNA network in burns and keloids and provided a potentially novel perspective on the DELs-DEMis-DEMs ceRNA network, contributing to the elucidation of the ceRNA regulatory mechanisms in the pathogenesis of burns and keloids. Nevertheless, systematic and rigorous experimental validations are indispensable to confirm our findings.
Collapse
Affiliation(s)
- Yueru Wang
- College of Medicine and Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi’an, Shaanxi, P.R. China
| | - Zhichao Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, Shaanxi, P.R. China
| | - Jiaojiao Pan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, Shaanxi, P.R. China
| | - He Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, Shaanxi, P.R. China
| | - Ziwen Lei
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, Shaanxi, P.R. China
| | - Jing Liu
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, Shaanxi, P.R. China
| | - Junbo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, Shaanxi, P.R. China
| | - Haizhen Lv
- Department of Pharmacy, Shaanxi Provincial Hospital of Tuberculosis Prevention and Treatment, Xi’an, P.R. China
| | - Fei Luan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, Shaanxi, P.R. China
| |
Collapse
|
2
|
Ye X, Zheng J, Hu D, Liu L, Chen F, Cai X, Xu Y, Li L, Lin J, Liu Q, Sun Y. Identification of increased dedifferentiation along the Prom1+ cancer cells in Müllerian adenosarcoma with sarcomatous overgrowth. Br J Cancer 2025; 132:438-449. [PMID: 39920368 PMCID: PMC11876574 DOI: 10.1038/s41416-025-02943-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/08/2024] [Accepted: 01/14/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Müllerian adenosarcoma (MA) is a rare tumour accounts for 5-7% of uterine sarcomas. Tumours with sarcomatous overgrowth (MASO) or high-grade tend to be aggressive. However, the tumour aetiology is elusive. METHODS Single-cell RNA sequencing and bioinformatics were used to analyse the MASO and paired normal tissues. Expression and clinical significance of key genes were analysed by TCGA data and immunohistochemistry. In vitro experiment was used to verify the effect of E2F1 in cell dedifferentiation. RESULTS We prove malignant stromal cells originate from fibrous tissue, Prom1-derived with complex intra-tumoral heterogeneity. Along the developmental trajectory, we discover three phenotypes of Prom1+ cancer cells (differentiation-like, intermediate-like, dedifferentiation-like). A distinct HMGB2/3+ subtype of Prom1+ cluster is predominant dedifferentiation-like cancer cells, with high proliferation and stemness traits at the tail of trajectory. E2F1 is a master transcription factor for Prom1 lineage dedifferentiation, which could occupy the HMGB2/3 promoter to enhance their transcription, facilitating the stemness and self-renewal of cancer cells. Gene signature of Prom1 lineage is associated with poorer prognosis in uterine malignancies. The expression of Prom1 and HMGB3 was verified by immunohistochemistry. CONCLUSIONS Our study reveal the heterogeneity and dynamics of Prom1 lineage cells, which are key to tailor efficient therapies for MASO.
Collapse
Affiliation(s)
- Xingming Ye
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jianfeng Zheng
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Dan Hu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Li Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Fukun Chen
- Geneplus-Beijing Institute, Beijing, China
| | - Xintong Cai
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yangmei Xu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lifeng Li
- Geneplus-Beijing Institute, Beijing, China
| | - Jie Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yang Sun
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| |
Collapse
|
3
|
Kim HJ, Kim YH. Comprehensive Insights into Keloid Pathogenesis and Advanced Therapeutic Strategies. Int J Mol Sci 2024; 25:8776. [PMID: 39201463 PMCID: PMC11354446 DOI: 10.3390/ijms25168776] [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: 06/26/2024] [Revised: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 09/02/2024] Open
Abstract
Keloid scars, characterized by abnormal fibroproliferation and excessive extracellular matrix (ECM) production that extends beyond the original wound, often cause pruritus, pain, and hyperpigmentation, significantly impacting the quality of life. Keloid pathogenesis is multifactorial, involving genetic predisposition, immune response dysregulation, and aberrant wound-healing processes. Central molecular pathways such as TGF-β/Smad and JAK/STAT are important in keloid formation by sustaining fibroblast activation and ECM deposition. Conventional treatments, including surgical excision, radiation, laser therapies, and intralesional injections, yield variable success but are limited by high recurrence rates and potential adverse effects. Emerging therapies targeting specific immune pathways, small molecule inhibitors, RNA interference, and mesenchymal stem cells show promise in disrupting the underlying mechanisms of keloid pathogenesis, potentially offering more effective and lasting treatment outcomes. Despite advancements, further research is essential to fully elucidate the precise mechanisms of keloid formation and to develop targeted therapies. Ongoing clinical trials and research efforts are vital for translating these scientific insights into practical treatments that can markedly enhance the quality of life for individuals affected by keloid scars.
Collapse
Affiliation(s)
- Hyun Jee Kim
- Department of Dermatology, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea;
| | - Yeong Ho Kim
- Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
4
|
Gao R, Mao J. Noncoding RNA-Mediated Epigenetic Regulation in Hepatic Stellate Cells of Liver Fibrosis. Noncoding RNA 2024; 10:44. [PMID: 39195573 DOI: 10.3390/ncrna10040044] [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: 05/22/2024] [Revised: 07/09/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
Liver fibrosis is a significant contributor to liver-related disease mortality on a global scale. Despite this, there remains a dearth of effective therapeutic interventions capable of reversing this condition. Consequently, it is imperative that we gain a comprehensive understanding of the underlying mechanisms driving liver fibrosis. In this regard, the activation of hepatic stellate cells (HSCs) is recognized as a pivotal factor in the development and progression of liver fibrosis. The role of noncoding RNAs (ncRNAs) in epigenetic regulation of HSCs transdifferentiation into myofibroblasts has been established, providing new insights into gene expression changes during HSCs activation. NcRNAs play a crucial role in mediating the epigenetics of HSCs, serving as novel regulators in the pathogenesis of liver fibrosis. As research on epigenetics expands, the connection between ncRNAs involved in HSCs activation and epigenetic mechanisms becomes more evident. These changes in gene regulation have attracted considerable attention from researchers in the field. Furthermore, epigenetics has contributed valuable insights to drug discovery and the identification of therapeutic targets for individuals suffering from liver fibrosis and cirrhosis. As such, this review offers a thorough discussion on the role of ncRNAs in the HSCs activation of liver fibrosis.
Collapse
Affiliation(s)
- Ruoyu Gao
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jingwei Mao
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
5
|
Jia H, Chang Y, Chen Y, Chen X, Zhang H, Hua X, Xu M, Sheng Y, Zhang N, Cui H, Han L, Zhang J, Fu X, Song J. A single-cell atlas of lung homeostasis reveals dynamic changes during development and aging. Commun Biol 2024; 7:427. [PMID: 38589700 PMCID: PMC11001898 DOI: 10.1038/s42003-024-06111-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: 08/08/2023] [Accepted: 03/26/2024] [Indexed: 04/10/2024] Open
Abstract
Aging is a global challenge, marked in the lungs by function decline and structural disorders, which affects the health of the elderly population. To explore anti-aging strategies, we develop a dynamic atlas covering 45 cell types in human lungs, spanning from embryonic development to aging. We aim to apply the discoveries of lung's development to address aging-related issues. We observe that both epithelial and immune cells undergo a process of acquisition and loss of essential function as they transition from development to aging. During aging, we identify cellular phenotypic alternations that result in reduced pulmonary compliance and compromised immune homeostasis. Furthermore, we find a distinctive expression pattern of the ferritin light chain (FTL) gene, which increases during development but decreases in various types of lung cells during the aging process.
Collapse
Affiliation(s)
- Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yulin Chen
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xiao Chen
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hang Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengda Xu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixuan Sheng
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Zhang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Cui
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Han
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of General Surgery, Yanan Hospital, Kunming Medical University, Kunming, China
| | - Jian Zhang
- Thoracic Surgery Department, the third affiliated hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China.
| | - Xiaodong Fu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Su L, Han J. Non-coding RNAs in hypertrophic scars and keloids: Current research and clinical relevance: A review. Int J Biol Macromol 2024; 256:128334. [PMID: 38007032 DOI: 10.1016/j.ijbiomac.2023.128334] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/28/2023] [Accepted: 11/12/2023] [Indexed: 11/27/2023]
Abstract
Hypertrophic scars (HS) and keloids (KD) are lesions that develop as a result of excessive fibroblast proliferation and collagen deposition in response to dermal injury, leading to dysregulation of the inflammatory, proliferative, and remodeling phases during wound healing. HS and KD affect up to 90 % of the population and are associated with lower quality of life, physical health, and mental status in patients. Efficient targeted treatment represents a significant challenge, primarily due to our limited understanding of their underlying pathogenesis. Non-coding RNAs (ncRNAs), which constitute a significant portion of the human transcriptome with minimal or no protein-coding capacity, have been implicated in various cellular physiologies and pathologies and may serve as diagnostic indicators or therapeutic targets. NcRNAs have been found to be aberrantly expressed and regulated in HS and KD. This review provides a summary of the expression profiles and molecular mechanisms of three common ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in HS and KD. It also discusses their potential as biomarkers for the diagnosis and treatment of these diseases and provides novel insights into epigenetic-based diagnosis and treatment strategies for HS and KD.
Collapse
Affiliation(s)
- Linlin Su
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
7
|
Ma Y, Liu Z, Miao L, Jiang X, Ruan H, Xuan R, Xu S. Mechanisms underlying pathological scarring by fibroblasts during wound healing. Int Wound J 2023. [PMID: 36726192 DOI: 10.1111/iwj.14097] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Pathological scarring is an abnormal outcome of wound healing, which often manifests as excessive proliferation and transdifferentiation of fibroblasts (FBs), and excessive deposition of the extracellular matrix. FBs are the most important effector cells involved in wound healing and scar formation. The factors that promote pathological scar formation often act on the proliferation and function of FB. In this study, we describe the factors that lead to abnormal FB formation in pathological scarring in terms of the microenvironment, signalling pathways, epigenetics, and autophagy. These findings suggest that understanding the causes of abnormal FB formation may aid in the development of precise and effective preventive and treatment strategies for pathological scarring that are associated with improved quality of life of patients.
Collapse
Affiliation(s)
- Yizhao Ma
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Zhifang Liu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - LinLin Miao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Xinyu Jiang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Hongyu Ruan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Rongrong Xuan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Suling Xu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
8
|
Lee CC, Tsai CH, Chen CH, Yeh YC, Chung WH, Chen CB. An updated review of the immunological mechanisms of keloid scars. Front Immunol 2023; 14:1117630. [PMID: 37033989 PMCID: PMC10075205 DOI: 10.3389/fimmu.2023.1117630] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Keloid is a type of disfiguring pathological scarring unique to human skin. The disorder is characterized by excessive collagen deposition. Immune cell infiltration is a hallmark of both normal and pathological tissue repair. However, the immunopathological mechanisms of keloid remain unclear. Recent studies have uncovered the pivotal role of both innate and adaptive immunity in modulating the aberrant behavior of keloid fibroblasts. Several novel therapeutics attempting to restore regulation of the immune microenvironment have shown variable efficacy. We review the current understanding of keloid immunopathogenesis and highlight the potential roles of immune pathway-specific therapeutics.
Collapse
Affiliation(s)
- Chih-Chun Lee
- 1 Department of Medical Education, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chia-Hsuan Tsai
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Hao Chen
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yuan-Chieh Yeh
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Hung Chung
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taiwan
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Taipei, Taiwan
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Linkou, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chun-Bing Chen
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taiwan
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Taipei, Taiwan
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Linkou, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- *Correspondence: Chun-Bing Chen, ;
| |
Collapse
|
9
|
The Intricate Interplay between the ZNF217 Oncogene and Epigenetic Processes Shapes Tumor Progression. Cancers (Basel) 2022; 14:cancers14246043. [PMID: 36551531 PMCID: PMC9776013 DOI: 10.3390/cancers14246043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
The oncogenic transcription factor ZNF217 orchestrates several molecular signaling networks to reprogram integrated circuits governing hallmark capabilities within cancer cells. High levels of ZNF217 expression provide advantages to a specific subset of cancer cells to reprogram tumor progression, drug resistance and cancer cell plasticity. ZNF217 expression level, thus, provides a powerful biomarker of poor prognosis and a predictive biomarker for anticancer therapies. Cancer epigenetic mechanisms are well known to support the acquisition of hallmark characteristics during oncogenesis. However, the complex interactions between ZNF217 and epigenetic processes have been poorly appreciated. Deregulated DNA methylation status at ZNF217 locus or an intricate cross-talk between ZNF217 and noncoding RNA networks could explain aberrant ZNF217 expression levels in a cancer cell context. On the other hand, the ZNF217 protein controls gene expression signatures and molecular signaling for tumor progression by tuning DNA methylation status at key promoters by interfering with noncoding RNAs or by refining the epitranscriptome. Altogether, this review focuses on the recent advances in the understanding of ZNF217 collaboration with epigenetics processes to orchestrate oncogenesis. We also discuss the exciting burgeoning translational medicine and candidate therapeutic strategies emerging from those recent findings connecting ZNF217 to epigenetic deregulation in cancer.
Collapse
|
10
|
Epigenetic Dysregulation in Autoimmune and Inflammatory Skin Diseases. Clin Rev Allergy Immunol 2022; 63:447-471. [DOI: 10.1007/s12016-022-08956-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2022] [Indexed: 11/11/2022]
|
11
|
Liu S, Yang H, Song J, Zhang Y, Abualhssain ATH, Yang B. Keloid: Genetic susceptibility and contributions of genetics and epigenetics to its pathogenesis. Exp Dermatol 2022; 31:1665-1675. [PMID: 36052657 DOI: 10.1111/exd.14671] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/29/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Keloid, characterized by fibroproliferative disorders of the skin, can be developed in people of different genders, ages, and ethnicities. Keloid can appear in any part of the body but are especially common on the earlobe, upper torso, and triangular muscle. The genetic heterogeneity and susceptibility of KD (keloid) vary among different races and ethnicities. Studies have found that multiple loci on multiple chromosomes are associated with the pathogenesis of KD, and specific gene variants may also be involved. Despite multiple investigations attempting to uncover the etiology of keloid formation, the genetic mechanism of keloid formation remains unknown. To establish a foundation for a better understanding of the genetics and epigenetics of keloids, we have evaluated and summarized current studies which are mostly related to heredity, genetic polymorphisms, predisposing gene, DNA methylation, and non-coding RNA. We also discussed the problems and potential of genetic and epigenetic investigations of keloids, with the goal of developing new therapeutic approaches to enhance the prognosis of keloid patients.
Collapse
Affiliation(s)
- Shuangfei Liu
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Huan Yang
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Jinru Song
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yue Zhang
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | | | - Bin Yang
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
LncRNA GNAS-AS1 knockdown inhibits keloid cells growth by mediating the miR-188-5p/RUNX2 axis. Mol Cell Biochem 2022; 478:707-719. [PMID: 36036334 DOI: 10.1007/s11010-022-04538-6] [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: 06/15/2021] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Keloid is a common dermis tumor, occurring repeatedly, affecting the quality of patients' life. Long non-coding RNAs (lncRNAs) have crucial regulatory capacities in skin scarring formation and subsequent scar carcinogenesis. The intention of this study was to investigate the mechanism and function of GNAS antisense-1 (GNAS-AS1) in keloids. Clinical samples were collected to evaluate the expression of GNAS-AS1, RUNX2, and miR-188-5p by qRT-PCR. The proliferation, migration, and invasion of HKF cells were detected by CCK-8, wound healing, and Transwell assays. The expression levels of mRNA and protein were examined through qRT-PCR and Western blot assay. Luciferase reporter assay was used to identify the binding relationship among GNAS-AS1, miR-188-5p, and Runt-related transcription factor 2 (RUNX2). GNAS-AS1 and RUNX2 expressions were remarkably enhanced, and miR-188-5p expression was decreased in keloid clinical tissues and HKF cells. GNAS-AS1 overexpression promoted cells proliferation, migration, and invasion, while GNAS-AS1 knockdown had the opposite trend. Furthermore, overexpression of GNAS-AS1 reversed the inhibitory effect of 5-FU on cell proliferation, migration, and invasion. MiR-188-5p inhibition or RUNX2 overexpression could enhance the proliferation, migration, and invasion of HKF cells. GNAS-AS1 targeted miR-188-5p to regulate RUNX2 expression. In addition, the inhibition effects of GNAS-AS1 knockdown on HKF cells could be reversed by inhibition of miR-188-5p or overexpression of RUNX2, while RUNX2 overexpression eliminated the suppressive efficaciousness of miR-188-5p mimics on HKF cells growth. GNAS-AS1 knockdown could regulate the miR-188-5p/RUNX2 signaling axis to inhibit the growth and migration in keloid cells. It is suggested that GNAS-AS1 may become a new target for the prevention and treatment of keloid.
Collapse
|
13
|
Yu X, Zhu X, Xu H, Li L. Emerging roles of long non-coding RNAs in keloids. Front Cell Dev Biol 2022; 10:963524. [PMID: 36046343 PMCID: PMC9421354 DOI: 10.3389/fcell.2022.963524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022] Open
Abstract
Keloids are pathologic wound healing conditions caused by fibroblast hyperproliferation and excess collagen deposition following skin injury or irritation, which significantly impact patients by causing psychosocial and functional distress. Extracellular matrix (ECM) deposition and human fibroblast proliferation represents the main pathophysiology of keloid. Long non-coding RNAs (LncRNAs) play important roles in many biological and pathological processes, including development, differentiation and carcinogenesis. Recently, accumulating evidences have demonstrated that deregulated lncRNAs contribute to keloids formation. The present review summarizes the researches of deregulated lncRNAs in keloid. Exploring lncRNA-based methods hold promise as new effective therapies against keloid.
Collapse
|
14
|
Chen Y, Chen C, Fang J, Su K, Yuan Q, Hou H, Xin H, Sun J, Huang C, Li S, Yuan Z, Luo S. Targeting the Akt/PI3K/mTOR signaling pathway for complete eradication of keloid disease by sunitinib. Apoptosis 2022; 27:812-824. [PMID: 35802302 DOI: 10.1007/s10495-022-01744-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2022] [Indexed: 11/24/2022]
Abstract
Keloid disease is a nodular lesion, tumor-like but not cancerous, and characterized of excessive proliferation of fibroblasts and deposition of extracellular matrix (ECM) components. This condition often causes itching, pain and cosmetic disfigurement, significantly reducing patient quality of life. To date, no universally effective therapies are available, possibly due to inadequate understanding of keloid pathogenesis. As an oral small-molecule inhibitor of certain tyrosine kinase receptors, sunitinib has shown significant therapeutic effects in renal cell carcinoma (RCC) and gastrointestinal stromal tumor (GIST). However, it has never been tested if keloid therapy can be effective for the management of keloids. This study thus aims to explore the potential of sunitinib for keloid treatment. Keloid-derived fibroblasts (KFs) were successfully isolated and demonstrated proliferative advantage to normal skin-derived fibroblasts (NFs). Additionally, sunitinib showed specific cytotoxicity and inhibition of invasion, and induced cell cycle arrest and significant apoptosis in KFs. These effects were accompanied by complete suppression of ECM component expression, including collagen types 1 and 3, upregulation of autophagy-associated LC3B and significant suppression of the Akt/PI3K/mTOR pathway. Moreover, a keloid explant culture model was successfully established and used to test the therapeutic efficacy of sunitinib on keloid formation in nude mice. Sunitinib was found to induce complete regression of keloid explant fragments in nude mice, showing significantly higher therapeutic efficacy than the most commonly used intralesional drug triamcinolone acetonide (TAC). These data suggest that sunitinib effectively inhibits keloid development through suppression of the Akt/PI3K/mTOR pathway and thus can be potentially developed as a monotherapy or combination therapy for the effective treatment of keloid disease.
Collapse
Affiliation(s)
- Yiqing Chen
- The Second School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, China.,Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China
| | - Chunlin Chen
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China
| | - Junren Fang
- The Second School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, China.,Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China
| | - Kui Su
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Qian Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Huan Hou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Huijuan Xin
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, 510317 Guangzhou, China
| | - Jianwu Sun
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Chaohong Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Shuyi Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Zhengqiang Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China.
| | - Shengkang Luo
- The Second School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, China. .,Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China.
| |
Collapse
|
15
|
Long non-coding RNA HOXA11-AS contributes to the formation of keloid by relieving the inhibition of miR-182-5p on ZNF217. Burns 2022:S0305-4179(22)00192-9. [DOI: 10.1016/j.burns.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2022]
|
16
|
Xia Y, Wang Y, Shan M, Hao Y, Liu H, Chen Q, Liang Z. Advances in the pathogenesis and clinical application prospects of tumor biomolecules in keloid. BURNS & TRAUMA 2022; 10:tkac025. [PMID: 35769828 PMCID: PMC9233200 DOI: 10.1093/burnst/tkac025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/13/2022] [Indexed: 12/29/2022]
Abstract
Keloid scarring is a kind of pathological healing manifestation after skin injury and possesses various tumor properties, such as the Warburg effect, epithelial-mesenchymal transition (EMT), expression imbalances of apoptosis-related genes and the presence of stem cells. Abnormal expression of tumor signatures is critical to the initiation and operation of these effects. Although previous experimental studies have recognized the potential value of a single or several tumor biomolecules in keloids, a comprehensive evaluation system for multiple tumor signatures in keloid scarring is still lacking. This paper aims to summarize tumor biomolecules in keloids from the perspectives of liquid biopsy, genetics, proteomics and epigenetics and to investigate their mechanisms of action and feasibility from bench to bedside. Liquid biopsy is suitable for the early screening of people with keloids due to its noninvasive and accurate performance. Epigenetic biomarkers do not require changes in the gene sequence and their reversibility and tissue specificity make them ideal therapeutic targets. Nonetheless, given the ethnic specificity and genetic predisposition of keloids, more large-sample multicenter studies are indispensable for determining the prevalence of these signatures and for establishing diagnostic criteria and therapeutic efficacy estimations based on these molecules.
Collapse
Affiliation(s)
- Yijun Xia
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Mengjie Shan
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Yan Hao
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Hao Liu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Qiao Chen
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Zhengyun Liang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
17
|
Amjadian S, Moradi S, Mohammadi P. The emerging therapeutic targets for scar management: genetic and epigenetic landscapes. Skin Pharmacol Physiol 2022; 35:247-265. [PMID: 35696989 PMCID: PMC9533440 DOI: 10.1159/000524990] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 04/22/2022] [Indexed: 11/28/2022]
Abstract
Background Wound healing is a complex process including hemostasis, inflammation, proliferation, and remodeling during which an orchestrated array of biological and molecular events occurs to promote skin regeneration. Abnormalities in each step of the wound healing process lead to reparative rather than regenerative responses, thereby driving the formation of cutaneous scar. Patients suffering from scars represent serious health problems such as contractures, functional and esthetic concerns as well as painful, thick, and itchy complications, which generally decrease the quality of life and impose high medical costs. Therefore, therapies reducing cutaneous scarring are necessary to improve patients' rehabilitation. Summary Current approaches to remove scars, including surgical and nonsurgical methods, are not efficient enough, which is in principle due to our limited knowledge about underlying mechanisms of pathological as well as the physiological wound healing process. Thus, therapeutic interventions focused on basic science including genetic and epigenetic knowledge are recently taken into consideration as promising approaches for scar management since they have the potential to provide targeted therapies and improve the conventional treatments as well as present opportunities for combination therapy. In this review, we highlight the recent advances in skin regenerative medicine through genetic and epigenetic approaches to achieve novel insights for the development of safe, efficient, and reproducible therapies and discuss promising approaches for scar management. Key Message Genetic and epigenetic regulatory switches are promising targets for scar management, provided the associated challenges are to be addressed.
Collapse
Affiliation(s)
- Sara Amjadian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Parvaneh Mohammadi
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- *Parvaneh Mohammadi,
| |
Collapse
|
18
|
Abstract
Significance: Keloid scarring is cosmetically disfiguring, psychosocially distressing, and can be physically disabling. The pathophysiology of keloid formation is poorly understood and subsequently, treatment options are ill defined, limited, and largely unsatisfactory. Therefore, in view of its unsatisfactory and recalcitrant management, keloid therapy is often seen as a financial burden affecting both patients and the health care systems. Recent Advances: Increased research on the genetic and epigenetic mechanisms in keloids has broadened our understanding of keloid pathobiology. Epigenetic mechanisms, mainly DNA methylation, histone modification, and noncoding RNAs, are currently being widely investigated. Advances in genetic sequencing technology and reduced cost have aided this endeavor. Studies on blood and patient-derived keloid tissue are being done with therapeutic agents targeting epigenetic and genetic markers with the shared goal of identifying the pathways underlying the initiation and maintenance of keloids. These advances have informed us of multiple complex molecular pathways implicated in keloids, which are yet to be fully elucidated. Critical Issues: Improved understanding of the genetic and epigenetic causes implicated in keloids will enhance our knowledge of this enigmatic disorder and likely lead to the development of therapeutic targets based on the available clinical and experimental studies. Due to the incomplete knowledge of molecular targets involved in keloid scarring pathways, therapeutics is still lagging for this clinically and scientifically important condition. Future Directions: Focused research on the identification of molecular targets and mechanistic pathways implicated in keloids is required to generate novel antifibrotic therapeutic options to decrease or eradicate recurrence of the disease as well as associated morbidity and improve the quality of life of those affected with keloids.
Collapse
Affiliation(s)
- Dennias Tonderai Nyika
- MRC Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla P. Khumalo
- MRC Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- MRC Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Plastic and Reconstructive Surgery Research, Wound Healing Unit, NIHR Manchester Biomedical Research, Centre for Dermatology Research, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
Chen Z, Chu X, Xu J. Detection and analysis of long noncoding RNA expression profiles related to epithelial-mesenchymal transition in keloids. Biomed Eng Online 2022; 21:2. [PMID: 35012558 PMCID: PMC8751032 DOI: 10.1186/s12938-022-00976-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/03/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The role of epithelial-mesenchymal transition (EMT) in the pathogenesis of keloids is currently raising increasing attention. Long noncoding RNAs (lncRNAs) govern a variety of biological processes, such as EMT, and their dysregulation is involved in many diseases including keloid disease. The aim of this study was to identify differentially expressed EMT-related lncRNAs in keloid tissues versus normal tissues and to interpret their functions. RESULTS Eleven lncRNAs and 16 mRNAs associated with EMT were identified to have differential expression between keloid and normal skin tissues (fold change > 1.5, P < 0.05). Gene Ontology (GO) analysis showed that these differentially expressed mRNAs functioned in the extracellular matrix, protein binding, the positive regulation of cellular processes, the Set1C/COMPASS complex and histone acetyltransferase activity. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that these mRNAs are involved in pathways in cancer. The lncRNA, XLOC_000587 may promote cell proliferation and migration by enhancing the expression of ENAH, while AF268386 may facilitate the invasive growth of keloids by upregulating DDR2. CONCLUSIONS We characterized the differential expression profiles of EMT-related lncRNAs and mRNAs in keloids, which may contribute to preventing the occurrence and development of keloids by targeting the corresponding signaling pathways. These lncRNAs and mRNAs may provide biomarkers for keloid diagnosis and serve as potential targets for the treatment of this disease.
Collapse
Affiliation(s)
- Zhixiong Chen
- Department of Plastic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Xi Chu
- Department of Plastic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Jinghong Xu
- Department of Plastic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
20
|
Wang Z, Jiao P, Zhong Y, Ji H, Zhang Y, Song H, Du H, Ding X, Wu H. The Endoplasmic Reticulum-Stressed Head and Neck Squamous Cell Carcinoma Cells Induced Exosomal miR-424-5p Inhibits Angiogenesis and Migration of Humanumbilical Vein Endothelial Cells Through LAMC1-Mediated Wnt/β-Catenin Signaling Pathway. Cell Transplant 2022; 31:9636897221083549. [PMID: 35315295 PMCID: PMC8943634 DOI: 10.1177/09636897221083549] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Under endoplasmic reticulum (ER) stress, tumor plays multifaceted roles in
endothelial cell dysfunction through secreting exosomal miRNAs. However, for the
head and neck squamous cell carcinoma (HNSCC), it is still unclear about the
impact of ER-stressed HNSCC cell derived exosomes on vascular endothelial cells.
To address this gap, herein, systemic research was conducted including isolation
and characterization of ER-stressed HNSCC cell (HN4 cell line as an in
vitro model) derived exosomes, identification of regulatory
exosomal miRNAs, target exploration and downstream signaling pathway
investigation of exosomal miRNAs in human umbilical vein endothelial cell
(HUVEC). ER-stressed HN4 cell-derived exosomes inhibited angiogenesis and
migration of HUVEC cells in vitro. Furthermore, RNA-seq
analysis demonstrated that miR-424-5p was highly upregulated in ER-stressed HN4
cell-derived exosomes. Through matrigel tube formation and transwell assays of
HUVEC cells, miR-424-5p displayed great capabilities on inhibiting angiogenesis
and migration. Finally, based on western blot and luciferase reporter, it was
demonstrated that LAMC1 is the target of miR-424-5p which could inhibit the
angiogenesis and migration of HUVEC cells by repressing the LAMC1-mediated
Wnt/β-catenin signaling pathway. ER-stressed HNSCC cell-induced exosomal
miR-424-5p inhibits angiogenesis and migration of HUVEC cells through
LAMC1-mediated Wnt/β-catenin signaling pathway. This study offers a new insight
for understanding the complicated mechanism behind ER-stress induced
anti-angiogenesis of HNSCC.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Pengfei Jiao
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Yi Zhong
- Department of General Dentistry, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Huan Ji
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Haiyang Song
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China.,Department of General Dentistry, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Hongming Du
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Ding
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Heming Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Policarpo R, d’Ydewalle C. Missing lnc(RNAs) in Alzheimer's Disease? Genes (Basel) 2021; 13:39. [PMID: 35052379 PMCID: PMC8774680 DOI: 10.3390/genes13010039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/26/2022] Open
Abstract
With the ongoing demographic shift towards increasingly elderly populations, it is estimated that approximately 150 million people will live with Alzheimer's disease (AD) by 2050. By then, AD will be one of the most burdensome diseases of this and potentially next centuries. Although its exact etiology remains elusive, both environmental and genetic factors play crucial roles in the mechanisms underlying AD neuropathology. Genome-wide association studies (GWAS) identified genetic variants associated with AD susceptibility in more than 40 different genomic loci. Most of these disease-associated variants reside in non-coding regions of the genome. In recent years, it has become clear that functionally active transcripts arise from these non-coding loci. One type of non-coding transcript, referred to as long non-coding RNAs (lncRNAs), gained significant attention due to their multiple roles in neurodevelopment, brain homeostasis, aging, and their dysregulation or dysfunction in neurological diseases including in AD. Here, we will summarize the current knowledge regarding genetic variations, expression profiles, as well as potential functions, diagnostic or therapeutic roles of lncRNAs in AD. We postulate that lncRNAs may represent the missing link in AD pathology and that unraveling their role may open avenues to better AD treatments.
Collapse
Affiliation(s)
- Rafaela Policarpo
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium;
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica N.V., 2340 Beerse, Belgium
| | - Constantin d’Ydewalle
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica N.V., 2340 Beerse, Belgium
| |
Collapse
|
22
|
Wu Q, Chen J, Tan Z, Wang D, Zhou J, Li D, Cen Y. Long non-coding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) regulates fibroblast growth factor receptor substrate 2 (FRS2) by targeting microRNA (miR)-29-3p in hypertrophic scar fibroblasts. Bioengineered 2021; 12:5210-5219. [PMID: 34414852 PMCID: PMC8806793 DOI: 10.1080/21655979.2021.1959221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play crucial roles in human diseases. However, the detailed role of lncRNAs in hypertrophic scar fibroblasts (HSFs) is inadequately understood. This study aimed to investigate the potential role of lncRNA nuclear enriched abundant transcript 1 (NEAT1) in hypertrophic scarring. Expression of lncRNAs, miRNAs, and genes were detected by polymerase chain reaction; protein expression was evaluated using western blotting. Cellular function was determined using the CCK-8 assay. The interaction between microRNA (miR)-29-3p and NEAT1 or fibroblast growth factor receptor substrate 2 (FRS2) was verified by luciferase and RNA pull-down assays. The results showed that NEAT1 was overexpressed in the hypertrophic dermis and in HSFs. However, knockdown of NEAT1 suppressed the proliferation and extracellular matrix (ECM) production of HSFs. Moreover, NEAT1 functioned as a competing endogenous RNA to upregulate FRS2 by sponging miR-29-3p. Downregulation of miR-29-3p or overexpression of FRS2 antagonized the effects of NEAT1 knockdown and promoted HSF proliferation and ECM release. In conclusion, NEAT1 knockdown protected against hypertrophic scarring by modulating the miR-29-3p/FRS2 axis, which is a viable target in scar treatment.
Collapse
Affiliation(s)
- Qinghua Wu
- The Department of Plastic and Burn Surgery of West China Hospital, Sichuan University, Chengdu, China.,Burn and Plastic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan
| | - Junjie Chen
- The Department of Plastic and Burn Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Ziming Tan
- Burn and Plastic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan
| | - Dehuai Wang
- Burn and Plastic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan
| | - Jianwen Zhou
- Burn and Plastic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan
| | - Dan Li
- Burn and Plastic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan
| | - Ying Cen
- The Department of Plastic and Burn Surgery of West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Li Y, Wu H, Wang Q, Xu S. ZNF217: the cerberus who fails to guard the gateway to lethal malignancy. Am J Cancer Res 2021; 11:3378-3405. [PMID: 34354851 PMCID: PMC8332857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/14/2021] [Indexed: 06/13/2023] Open
Abstract
The aberrant expression of the zinc finger protein 217 (ZNF217) promotes multiple malignant phenotypes, such as replicative immortality, maintenance of proliferation, malignant heterogeneity, metastasis, and cell death resistance, via diverse mechanisms, including transcriptional activation, mRNA N6-methyladenosine (m6A) regulation, and protein interactions. The induction of these cellular processes by ZNF217 leads to therapeutic resistance and patients' poor outcomes. However, few ZNF217 related clinical applications or trials, have been reported. Moreover, looming observations about ZNF217 roles in m6A regulation and cancer immune response triggered significant attention while lacking critical evidence. Thus, in this review, we revisit the literature about ZNF217 and emphasize its importance as a prognostic biomarker for early prevention and as a therapeutic target.
Collapse
Affiliation(s)
- Yingpu Li
- Department of Breast Surgery, Harbin Medical University Cancer HospitalHarbin, China
| | - Hao Wu
- Sino-Russian Medical Research Center, Harbin Medical University Cancer HospitalHarbin, China
- Heilongjiang Academy of Medical SciencesHarbin, China
| | - Qin Wang
- Department of Breast Surgery, Harbin Medical University Cancer HospitalHarbin, China
- Sino-Russian Medical Research Center, Harbin Medical University Cancer HospitalHarbin, China
- Heilongjiang Academy of Medical SciencesHarbin, China
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer HospitalHarbin, China
- Sino-Russian Medical Research Center, Harbin Medical University Cancer HospitalHarbin, China
- Heilongjiang Academy of Medical SciencesHarbin, China
| |
Collapse
|
24
|
Xu Q, Cheng D, Liu Y, Pan H, Li G, Li P, Li Y, Sun W, Ma D, Ni C. LncRNA-ATB regulates epithelial-mesenchymal transition progression in pulmonary fibrosis via sponging miR-29b-2-5p and miR-34c-3p. J Cell Mol Med 2021; 25:7294-7306. [PMID: 34180127 PMCID: PMC8335671 DOI: 10.1111/jcmm.16758] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/29/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Dysregulation of non‐coding RNAs (ncRNAs) has been proved to play pivotal roles in epithelial‐mesenchymal transition (EMT) and fibrosis. We have previously demonstrated the crucial function of long non‐coding RNA (lncRNA) ATB in silica‐induced pulmonary fibrosis‐related EMT progression. However, the underlying molecular mechanism has not been fully elucidated. Here, we verified miR‐29b‐2‐5p and miR‐34c‐3p as two vital downstream targets of lncRNA‐ATB. As opposed to lncRNA‐ATB, a significant reduction of both miR‐29b‐2‐5p and miR‐34c‐3p was observed in lung epithelial cells treated with TGF‐β1 and a murine silicosis model. Overexpression miR‐29b‐2‐5p or miR‐34c‐3p inhibited EMT process and abrogated the pro‐fibrotic effects of lncRNA‐ATB in vitro. Further, the ectopic expression of miR‐29b‐2‐5p and miR‐34c‐3p with chemotherapy attenuated silica‐induced pulmonary fibrosis in vivo. Mechanistically, TGF‐β1‐induced lncRNA‐ATB accelerated EMT as a sponge of miR‐29b‐2‐5p and miR‐34c‐3p and shared miRNA response elements with MEKK2 and NOTCH2, thus relieving these two molecules from miRNA‐mediated translational repression. Interestingly, the co‐transfection of miR‐29b‐2‐5p and miR‐34c‐3p showed a synergistic suppression effect on EMT in vitro. Furthermore, the co‐expression of these two miRNAs by using adeno‐associated virus (AAV) better alleviated silica‐induced fibrogenesis than single miRNA. Approaches aiming at lncRNA‐ATB and its downstream effectors may represent new effective therapeutic strategies in pulmonary fibrosis.
Collapse
Affiliation(s)
- Qi Xu
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Demin Cheng
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yi Liu
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Honghong Pan
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Guanru Li
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ping Li
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yan Li
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenqing Sun
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Dongyu Ma
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chunhui Ni
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Zhang Y, Wang Q, Xu Y, Sun J, Ding Y, Wang L, Chen B, Sun K, Chen J. Mitomycin C Inhibits Esophageal Fibrosis by Regulating Cell Apoptosis and Autophagy via lncRNA-ATB and miR-200b. Front Mol Biosci 2021; 8:675757. [PMID: 34079820 PMCID: PMC8165251 DOI: 10.3389/fmolb.2021.675757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/27/2021] [Indexed: 01/11/2023] Open
Abstract
Benign esophageal strictures (BESs) frequently results from esophageal fibrosis. The transformation of fibroblasts into fibrocyte is an important cause of fibrosis. The treatment of fibrosis is challenging. Some previous studies have indicated the antifibrotic effect of mitomycin C (MMC). However, the mechanism of action of MMC and its optimal dose for treatment remains unclear. In the present study, the role of MMC in fighting fibrosis and its mechanism was investigated. Human esophageal fibroblast cells (HEFs)were treated without or with MMC, at 2, 5, 10 μg/ml, combining with mimic lncRNA-ATB, miR-200b inhibitor, rapamycin (RAPA), and 3-Methyladenine (3-MA). The cell viability, and cell apoptosis were evaluated. In addition, expression of apoptosis related proteins (caspase8 and caspase3), autophagy related proteins (LC3II and ATG5) and fibrosis related proteins (α-SMA collagen-1 and TGF-β) were also evaluated. Furthermore, autophagosome was observed by transmission electron microscope. Results showed that the expression of lncRNA-ATB was down-regulated and miR-200b was up-regulated after treated with MMC. And MMC induced cell apoptosis and inhibited cell autophagy. On the other hand, RAPA, mimic lncRNA-ATB and miR-200b inhibitor reduced fibrogenic effect of MMC on HEFs. Collectively, this study suggests that MMC inhibited esophageal fibrosis by regulating cell apoptosis and autophagy via downregulating lncRNA-ATB and upregulating miR-200b.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Qinge Wang
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Yuping Xu
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Jing Sun
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Yanbo Ding
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Li Wang
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Bingfang Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Kewen Sun
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| | - Jianping Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- The First People's Hospital of Changzhou, Changzhou, China
| |
Collapse
|
26
|
Lv W, Ren Y, Hou K, Hu W, Yi Y, Xiong M, Wu M, Wu Y, Zhang Q. Epigenetic modification mechanisms involved in keloid: current status and prospect. Clin Epigenetics 2020; 12:183. [PMID: 33243301 PMCID: PMC7690154 DOI: 10.1186/s13148-020-00981-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/17/2020] [Indexed: 12/18/2022] Open
Abstract
Keloid, a common dermal fibroproliferative disorder, is benign skin tumors characterized by the aggressive fibroblasts proliferation and excessive accumulation of extracellular matrix. However, common therapeutic approaches of keloid have limited effectiveness, emphasizing the momentousness of developing innovative mechanisms and therapeutic strategies. Epigenetics, representing the potential link of complex interactions between genetics and external risk factors, is currently under intense scrutiny. Accumulating evidence has demonstrated that multiple diverse and reversible epigenetic modifications, represented by DNA methylation, histone modification, and non-coding RNAs (ncRNAs), play a critical role in gene regulation and downstream fibroblastic function in keloid. Importantly, abnormal epigenetic modification manipulates multiple behaviors of keloid-derived fibroblasts, which served as the main cellular components in keloid skin tissue, including proliferation, migration, apoptosis, and differentiation. Here, we have reviewed and summarized the present available clinical and experimental studies to deeply investigate the expression profiles and clarify the mechanisms of epigenetic modification in the progression of keloid, mainly including DNA methylation, histone modification, and ncRNAs (miRNA, lncRNA, and circRNA). Besides, we also provide the challenges and future perspectives associated with epigenetics modification in keloid. Deciphering the complicated epigenetic modification in keloid is hopeful to bring novel insights into the pathogenesis etiology and diagnostic/therapeutic targets in keloid, laying a foundation for optimal keloid ending.
Collapse
Affiliation(s)
- Wenchang Lv
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China
| | - Yuping Ren
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China
| | - Kai Hou
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China
| | - Weijie Hu
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China
| | - Yi Yi
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China
| | - Mingchen Xiong
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China
| | - Min Wu
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China.
| | - Yiping Wu
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China.
| | - Qi Zhang
- Department of Plastic and Aesthetic Surgery, NO 1095 Jiefang Avenue, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430000, Hubei, China.
| |
Collapse
|
27
|
LncRNA MALAT1 promotes wound healing via regulating miR-141-3p/ZNF217 axis. Regen Ther 2020; 15:202-209. [PMID: 33426220 PMCID: PMC7770423 DOI: 10.1016/j.reth.2020.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/16/2020] [Accepted: 09/25/2020] [Indexed: 01/22/2023] Open
Abstract
Background The process of wound healing is complex. Increasing evidences have shown that lncRNA MALAT1 is abundant in fibroblasts and may be engaged in wound healing process. Therefore, we explored the mechanism of MALAT1 affecting wound healing. Methods The expression levels of MALAT1, miR-141-3p as well as ZNF217 in human fibroblast cells (HFF-1) were quantified by qRT-PCR. HFF-1 proliferation was measured by MTT, while migration was detected by wound healing assay. SMAD2 activation and matrix proteins expression were detected by western blotting. The interaction between miR-141-3p and MALAT1 or ZNF217 was further confirmed using the luciferase reporter gene assay. In vivo wound healing was assessed by full-thickness wound healing model on C57BL/6 mice. Result Knockdown of MALAT1 as well as overexpression miR-141-3p remarkably inhibited the proliferation, migration and matrix protein expression in HFF-1 cells. MALAT1 directly targeted and inhibited the expression of miR-141-3p. MiR-141-3p suppressed the activation of TGF-β2/SMAD2 signaling pathway by targeting ZNF217. Knockdown of MALAT1 inhibited wound healing process in mice. Conclusions MALAT1 up-regulates ZNF217 expression by targeting miR-141-3p, thus enhances the activity of TGF-β2/SMAD2 signaling pathway and promotes wound healing process. This investigation shed new light on the understanding of the role of MALAT1 in wound healing, and may provide potential target for the diagnosis or therapy of chronic wounds.
Collapse
Key Words
- ECM, extra cellular matrix
- ELISA, enzyme linked immunosorbent assay
- EMT, epithelial mesenchymal transition
- HFF-1, human fibroblast cells
- MALAT1
- MALAT1, metastasis-associated lung adenocarcinoma transcript 1
- MTT, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide
- PVDF, polyvinylidene fluoride
- SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- TGF-β2, Transforming Growth Factor-β2
- Wound healing
- ZEB1, E-box binding homeobox 1
- ZNF217
- ZNF217, zinc-finger protein 217
- lncRNA, long non-coding RNA
- miR-141-3p
- qRT-PCR, quantitative real-time PCR
Collapse
|
28
|
Yin JL, Wu Y, Yuan ZW, Gao XH, Chen HD. Advances in scarless foetal wound healing and prospects for scar reduction in adults. Cell Prolif 2020; 53:e12916. [PMID: 33058377 PMCID: PMC7653265 DOI: 10.1111/cpr.12916] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/25/2020] [Accepted: 09/06/2020] [Indexed: 02/06/2023] Open
Abstract
Healing after mammalian skin injury involves the interaction between numerous cellular constituents and regulatory factors, which together form three overlapping phases: an inflammatory response, a proliferation phase and a remodelling phase. Any slight variation in these three stages can substantially alter the healing process and resultant production of scars. Of particular significance are the mechanisms responsible for the scar‐free phenomenon observed in the foetus. Uncovering such mechanisms would offer great expectations in the treatment of scars and therefore represents an important area of investigation. In this review, we provide a comprehensive summary of studies on injury‐induced skin regeneration within the foetus. The information contained in these studies provides an opportunity for new insights into the treatment of clinical scars based on the cellular and molecular processes involved.
Collapse
Affiliation(s)
- Jia-Li Yin
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yan Wu
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zheng-Wei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - Xing-Hua Gao
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hong-Duo Chen
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
29
|
Papoutsoglou P, Moustakas A. Long non-coding RNAs and TGF-β signaling in cancer. Cancer Sci 2020; 111:2672-2681. [PMID: 32485023 PMCID: PMC7419046 DOI: 10.1111/cas.14509] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is driven by genetic mutations in oncogenes and tumor suppressor genes and by cellular events that develop a misregulated molecular microenvironment in the growing tumor tissue. The tumor microenvironment is guided by the excessive action of specific cytokines including transforming growth factor-β (TGF-β), which normally controls embryonic development and the homeostasis of young or adult tissues. As a consequence of the genetic alterations generating a given tumor, TGF-β can preserve its homeostatic function and attempt to limit neoplastic expansion, whereas, once the tumor has progressed to an aggressive stage, TGF-β can synergize with various oncogenic stimuli to facilitate tumor invasiveness and metastasis. TGF-β signaling mechanisms via Smad proteins, various ubiquitin ligases, and protein kinases are relatively well understood. Such mechanisms regulate the expression of genes encoding proteins or non-coding RNAs. Among non-coding RNAs, much has been understood regarding the regulation and function of microRNAs, whereas the role of long non-coding RNAs is still emerging. This article emphasizes TGF-β signaling mechanisms leading to the regulation of non-coding genes, the function of such non-coding RNAs as regulators of TGF-β signaling, and the contribution of these mechanisms in specific hallmarks of cancer.
Collapse
Affiliation(s)
| | - Aristidis Moustakas
- Department of Medical Biochemistry and MicrobiologyScience for Life LaboratoryUppsala UniversityUppsalaSweden
| |
Collapse
|
30
|
LncRNA-ATB in cancers: what do we know so far? Mol Biol Rep 2020; 47:4077-4086. [PMID: 32248383 DOI: 10.1007/s11033-020-05415-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/30/2020] [Indexed: 01/06/2023]
Abstract
Cancer-related deaths did not apparently decrease in the past decades despite aggressive treatments. It's reported that cancer will become the leading cause of death worldwide in the twenty-first century. Increasing evidence has revealed that lncRNAs will emerge as promising cancer biomarkers or therapeutic targets in cancer treatment. LncRNA-ATB, a long noncoding RNA activated by TGF-β, was found to be abnormally expressed in certain cancers and participate in the development and progression of tumors. In addition, aberrant lncRNA-ATB expression was also associated with clinical characteristics of tumors. The purpose of this review is to summarize functions and underlying mechanisms of lncRNA-ATB in tumors, and discuss whether lncRNA-ATB can be a biomarker and therapeutic target in cancers.
Collapse
|
31
|
Duan X, Wu Y, Zhang Z, Lu Z. Identification and analysis of dysregulated lncRNA and associated ceRNA in the pathogenesis of keloid. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:222. [PMID: 32309369 PMCID: PMC7154391 DOI: 10.21037/atm.2020.01.07] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background Keloid is an excessive fibrosis disease caused by the abnormal proliferation of collagen fibers following trauma. Previous studies have shown that genetic factors have been considered to play important roles in keloid formation. This study is aimed to investigate the regulatory network of messenger RNAs (mRNAs) microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) in keloid, and identifying its key biomarkers. Methods We performed RNA-seq and miRNA-seq on keloid and normal skin samples. Sequencing datasets were analyzed by bioinformatics. Gene ontology (GO) and pathway analysis presented the characteristics of associated protein-coding genes. Differentially expressed ceRNAs were validated by quantitative reverse transcriptase-PCR (qRT-PCR). Results We identified a total of 319 lncRNAs, 1,533 mRNAs and 40 miRNAs as keloid-specific RNAs. Both the GO biological processes and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were analyzed for 1,219 specific genes with differentially expressed mRNAs. Then, with 509 key lncRNAs, 25 miRNAs, and 94 mRNAs, we constructed a ceRNA network and explored any potential underlying mechanisms. In the regulation of the actin cytokeleton pathway, we validated 2 pairs of ceRNAs EGFR/miR-370-3p/lnc-GLB1L-1 and ITGB5/ miR-204/ lnc-CASP9-3 in another sample size in keloid. Conclusions Through RNA-seq and miRNA-seq, we identified keloid-associated lncRNAs, mRNAs and miRNAs, which can be used as potential therapeutic targets and biomarkers for keloid. Our study may lay a foundation for future pathogenesis studies.
Collapse
Affiliation(s)
- Xilei Duan
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuemeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zheng Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhong Lu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
32
|
Liu P, Hu Y, Xia L, Du M, Hu Z. miR-4417 suppresses keloid fibrosis growth by inhibiting CyclinD1. J Biosci 2020. [DOI: 10.1007/s12038-020-0018-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
33
|
Expression profile and bioinformatics analyses of circular RNAs in keloid and normal dermal fibroblasts. Exp Cell Res 2020; 388:111799. [PMID: 31904383 DOI: 10.1016/j.yexcr.2019.111799] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/19/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022]
Abstract
Increasing evidence indicates that circular RNAs (circRNAs) play a crucial regulatory role in the pathogenesis of multiple diseases. However, no study has examined the potential biological function and expression profile of circRNAs in keloid dermal fibroblasts (KDFs). Therefore, the aim of this study to investigate the expression profile of circRNAs and analyze their role in KDFs. Bioinformatic analyses and high-throughput RNA sequencing technology were applied to explore the expression profile of circRNAs in 3 human KDFs and normal dermal fibroblasts (NDFs). The differentially expressed circRNAs were verified by reverse transcription PCR (RT-PCR), quantitative real-time-PCR (qRT-PCR) and Sanger sequencing. A circRNA-microRNA (miRNA)-mRNA interaction network was created using bioinformatics tools. Hsa_circ_0008259, was selected to confirm its function by qRT-PCR and Western blot. Collectively, 411 circRNAs, of which 206 were upregulated and 205 decreased, were found to be differentially expressed in KDFs and could bind to 2532 miRNA response elements (MREs). GO and KEGG pathways enrichment analyses showed that differentially expressed circRNAs were mainly involved in apoptosis, focal adhesion, PI3K-Akt and metabolic pathway, and may regulate the pathogenesis and development of keloid. Two candidate circRNAs (hsa_circRNA_0008259, hsa_circRNA_0005480) were verified to be significantly reduced in KDFs, and one candidate circRNA (hsa_circRNA_0002198) was significantly elevated in accordance with RNA-Seq data analysis. Overexpression of hsa_circRNA_0008259 inhibited type I and Ⅲ collagen expression. Taken together, our study demonstrates for the first time that circRNAs exhibits differential expression in KDFs, and may be key players in the pathogenesis of keloid, or act as biomarkers of keloid.
Collapse
|
34
|
Liu P, Hu Y, Xia L, DU M, Hu Z. miR-4417 suppresses keloid fibrosis growth by inhibiting CyclinD1. J Biosci 2020; 45:47. [PMID: 32345773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mounting evidence has reported that microRNAs (miRNAs) play irreplaceable roles in the development of keloid fibrosis. miR-4417 has been reported to contribute to nickel chloride-promoted lung epithelial cell fibrogenesis and tumorigenesis. However, whether miR-4417 is involved in keloid fibrogenesis as well as its underlying mechanisms remain largely elusive. In this study, the expression levels of miR-4417 and CyclinD1 in keloid tissues and fibroblasts were examined by qRT-PCR. Cell proliferation was determined by CCK assay. Western blot and flow cytometry were performed to evaluate cell apoptosis. Cell migration and invasion were measured by Transwell assay. Luciferase reporter assay was used to confirm the relationship between miR4417 and CyclinD1. As a result, we found that miR-4417 was significantly down-regulated in keloid tissues and fibroblasts. miR-4417 up-regulation led to the suppression of proliferation, migration, and invasion, while induced cell apoptosis in keloid fibroblasts. However, miR-4417 depletion exerted an opposite effect. CyclinD1 harbored the binding sites with miR-4417. Besides, the expression of CyclinD1 was evidently decreased in keloid tissues and fibroblasts. Meanwhile, miR-4417 was negatively correlated with CyclinD1 in keloid tissue. The effect of CyclinD1 knockdown on keloid fibroblasts was similar to that of miR-4417 overexpression. Furthermore, the elevated of CyclinD1 expression rescued the effect of miR-4417 up-regulation on keloid fibroblasts. miR-4417/CyclinD1 axis was required for cell proliferation, apoptosis, migration, and invasion in keloid fibroblasts. In conclusion, miR-4417 and CyclinD1 may be potential therapeutic targets for the treatment of keloid.
Collapse
Affiliation(s)
- Pei Liu
- Department of Plastic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | | | | | | | | |
Collapse
|
35
|
Zhu Z, Hou Q, Li M, Fu X. Molecular mechanism of myofibroblast formation and strategies for clinical drugs treatments in hypertrophic scars. J Cell Physiol 2019; 235:4109-4119. [PMID: 31612497 DOI: 10.1002/jcp.29302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022]
Abstract
Hypertrophic scars (HTS) commonly occurred after burn and trauma. It was characterized by the excessive deposition of extracellular matrix with the inadequate remodeling, which could result in severe physiological and psychological problems. However, the effective available prevention and treatment measures were still limited. The main pathological feature of HTS was the excessive formation of myofibroblasts, and they persist in the repaired tissue. To better understand the mechanics of this process, this review focused on the characteristics and formation of myofibroblasts, the main effector cells in HTS. We summarized the present theories and opinions on myofibroblasts formation from the perspective of related signaling pathways and epigenetic regulation, such as DNA methylation, miRNA/lncRNA/ceRNA action, histone modification, and so forth for a better understanding on the development of HTS. This information might assist in developing effective experimental and clinical treatment strategies. Additionally, we also summarized currently known clinical strategies for HTS treatment, including traditional drugs, molecular medicine, stem cells, and exosomes.
Collapse
Affiliation(s)
- Ziying Zhu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Qian Hou
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Meirong Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China.,Central Laboratory, Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya, China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
36
|
Yang Z, Jiang S, Shang J, Jiang Y, Dai Y, Xu B, Yu Y, Liang Z, Yang Y. LncRNA: Shedding light on mechanisms and opportunities in fibrosis and aging. Ageing Res Rev 2019; 52:17-31. [PMID: 30954650 DOI: 10.1016/j.arr.2019.04.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/24/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
Fibrosis is universally observed in multiple aging-related diseases and progressions and is characterized by excess accumulation of the extracellular matrix. Fibrosis occurs in various human organs and eventually results in organ failure. Noncoding RNAs (ncRNAs) have emerged as essential regulators of cellular signaling and relevant human diseases. In particular, the enigmatic class of long noncoding RNAs (lncRNAs) is a kind of noncoding RNA that is longer than 200 nucleotides and does not possess protein coding ability. LncRNAs have been identified to exert both promotive and inhibitory effects on the multifaceted processes of fibrosis. A growing body of studies has revealed that lncRNAs are involved in fibrosis in various organs, including the liver, heart, lung, and kidney. As lncRNAs have been increasingly identified, they have become promising targets for anti-fibrosis therapies. This review systematically highlights the recent advances regarding the roles of lncRNAs in fibrosis and sheds light on the use of lncRNAs as a potential treatment for fibrosis.
Collapse
|
37
|
Gao Z, Zhou H, Wang Y, Chen J, Ou Y. Regulatory effects of lncRNA ATB targeting miR-200c on proliferation and apoptosis of colorectal cancer cells. J Cell Biochem 2019; 121:332-343. [PMID: 31222825 DOI: 10.1002/jcb.29180] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 01/17/2023]
Abstract
This investigation was intended to elucidate whether long noncoding RNA (lncRNA)-activated by transforming growth factor-β (ATB) interacting with miR-200c could mediate colorectal cancer (CRC) progression, offering potential strategies for diagnosing and treating CRC. Here totally 315 patients with CRC were recruited, and their CRC tissues and adjacent normal tissues were gathered. Concurrently, four colon cancer cell lines (ie, SW620, Lovo, HCT116, and SW480) and the human colon mucosal epithelial cell line (NCM460) were also purchased. Moreover, si-ATB, si-NC, miR-200c mimic, miR-200c inhibitor, and miR-NC were prepared for transfection into the CRC cells, and their effects on CRC cell lines were evaluated based on the conduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, colony formation assay, and flow cytometry assay. Eventually, the Luciferase reporter gene assay was carried out to judge if there existed a targeted relationship between ATB and miR-200c. The results of Cox regression analyses suggested that overexpressed lncRNA ATB, underexpressed miR-200c, poor tumor differentiation, lymph-vascular invasion, and perineural invasion were symbolic of shortened survival of the patients with CRC (all P < .05). Besides, transfection of pcDNA3.1-ATB and miR-200c inhibitor could boost the viability and proliferation of Lovo and SW620 cell lines (all P < .05). Meanwhile, the expressions of p53 and p21 were also reduced under treatments of pcDNA3.1-ATB and miR-200c inhibitor (P < .05). In addition, CDK2 seemed to reverse the contribution of miR-200c to intensifying viability and proliferation of Lovo and SW420 cell lines (P < .05). Furthermore, ATB might downregulate miR-200c expression by targeting it (P < .05), and CDK2 was subjected to dual regulation of both ATB and miR-200c (P < .05). In conclusion, the lncRNA ATB/miR-200c/CDK2 signaling was responsible for intensified proliferation and prohibited apoptosis of CRC cells, which might provide effective approaches for diagnosing and treating CRC.
Collapse
Affiliation(s)
- Zhenyuan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Hairong Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yaping Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Juan Chen
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yimei Ou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
38
|
Zhu A, Sun Y, Ma Q, Xu F. Retracted
: lncRNA‐ATB promotes viability, migration, and angiogenesis in human microvascular endothelial cells by sponging microRNA‐195. J Cell Biochem 2019; 120:14360-14371. [DOI: 10.1002/jcb.28692] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/25/2018] [Accepted: 01/10/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Ai‐Dong Zhu
- Department of Vascular Surgery Jining No. 1 People's Hospital Jining China
- Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University Jining China
| | - Ying‐Ying Sun
- Department of Vascular Surgery Jining No. 1 People's Hospital Jining China
| | - Qiu‐Ju Ma
- Department of Vascular Surgery Jining No. 1 People's Hospital Jining China
| | - Fei Xu
- Department of Vascular Surgery Jining No. 1 People's Hospital Jining China
| |
Collapse
|
39
|
Huang C, Liu L, You Z, Du Y, Ogawa R. Managing keloid scars: From radiation therapy to actual and potential drug deliveries. Int Wound J 2019; 16:852-859. [PMID: 30864269 DOI: 10.1111/iwj.13104] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/07/2019] [Accepted: 02/10/2019] [Indexed: 12/19/2022] Open
Abstract
The aetiology of keloids is becoming clearer, but many questions remain, including about the most optimal treatment. Current therapies include surgical excision, radiotherapy, and various pharmaceutical drugs. However, none of these drugs are keloid-specific. Moreover, all current interventions are associated with high recurrence rates. Here, we review the pharmaceutical interventions that are currently available. All are based on the fact that keloids are an expanding solid mass with intense chronic inflammation at its advancing edges. Consequently, current pharmaceuticals aim to reduce the mass and/or symptoms of keloids, similar to surgery and radiotherapy. They include chemotherapies, immunotherapies, volume-reducing therapies, and anti-inflammatory therapies. We also describe new advances in keloid pharmaceuticals. They include drugs that were designed to treat systemic diseases such as hypertension or breast cancer but were found to also treat keloids. Furthermore, recent progress in genetic, epigenetic, and stem cell therapies suggests that they could become useful in the keloid field. This review of pharmaceutical advances will hopefully promote additional research and the development of effective and specific pharmaceuticals for keloids.
Collapse
Affiliation(s)
- Chenyu Huang
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Longwei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
40
|
Abstract
INTRODUCTION Keloids and hypertrophic scars are fibroproliferative disorders of the skin that result from abnormal healing of injured or irritated skin. Multiple studies suggest that genetic, systemic and local factors may contribute to the development and/or growth of keloids and hypertrophic scars. A key local factor may be mechanical stimuli. Here, we provide an up-to-date review of the studies on the roles that genetic variation, epigenetic modifications and mechanotransduction play in keloidogenesis. METHODS An English literature review was performed by searching the PubMed, Embase and Web of Science databases with the following keywords: genome-wide association study; epigenetics; non-coding RNA; microRNA; long non-coding RNA (lncRNA); DNA methylation; mechanobiology; and keloid. The searches targeted the time period between the date of database inception and July 2018. RESULTS Genetic studies identified several single-nucleotide polymorphisms and gene linkages that may contribute to keloid pathogenesis. Epigenetic modifications caused by non-coding RNAs (e.g. microRNAs and lncRNAs) and DNA methylation may also play important roles by inducing the persistent activation of keloidal fibroblasts. Mechanical forces and the ensuing cellular mechanotransduction may also influence the degree of scar formation, scar contracture and the formation/progression of keloids and hypertrophic scars. CONCLUSIONS Recent research indicates that the formation/growth of keloids and hypertrophic scars associate clearly with genetic, epigenetic, systemic and local risk factors, particularly skin tension around scars. Further research into scar-related genetics, epigenetics and mechanobiology may reveal molecular, cellular or tissue-level targets that could lead to the development of more effective prophylactic and therapeutic strategies for wounds/scars in the future.
Collapse
Affiliation(s)
- Chia-Hsuan Tsai
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Keelung & Chang Gung University College of Medicine, Taoyuan
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
41
|
Suppression of lncRNA-ATB prevents amyloid-β-induced neurotoxicity in PC12 cells via regulating miR-200/ZNF217 axis. Biomed Pharmacother 2018; 108:707-715. [DOI: 10.1016/j.biopha.2018.08.155] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 11/23/2022] Open
|
42
|
Chao L, Hua-Yu Z, Wen-Dong B, Mei S, Bin X, Da-Hai H, Yi L. miR-96 promotes collagen deposition in keloids by targeting Smad7. Exp Ther Med 2018; 17:773-781. [PMID: 30651862 PMCID: PMC6307430 DOI: 10.3892/etm.2018.7008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
The abnormal upregulation of transforming growth factor-β (TGF-β) signaling has been demonstrated to initiate keloid formation and progression. Keloid is a type of benign skin tumor that may occur following sustaining skin injury. microRNA-96 (miR-96) serves an important role in the progression of various malignant diseases. Using reverse transcription quantitative polymerase chain reaction (RT-qPCR), the present study demonstrated that miR-96 was overexpressed in keloid-derived fibroblasts (KFs). Luciferase reporter assay revealed mothers against decapentaplegic homolog (Smad)7, which is one of the important inhibitory factors in the TGF-β pathway, as a direct target of miR-96. miR-96 was initially observed to be correlated with the deposition of type I collagen in KFs in vitro. The miR-96 antagomir, was directly added into the keloid organ culture (OC) to find its significant antifibrotic potential, such as keloid OC shrinkage, exhibited by its dry weight loss and improved dermis architecture, exhibited by Masson's staining. Following miR-96 antagomir treatment, a reduction in the mRNA and protein expression levels of collagen type I α 1 chain and collagen type 3 α 1 chain within keloid OC tissues was observed. The present study revealed that miR-96 serves an important role in pathogenic keloid formation, suggesting that miR-96 antagomir has the potential to prevent keloid progression.
Collapse
Affiliation(s)
- Li Chao
- Burns and Plastic Surgery Center of People's Liberation Army, Lanzhou General Hospital of Chinese People's Liberation Army, Lanzhou, Gansu 730050, P.R. China
| | - Zhu Hua-Yu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Bai Wen-Dong
- Department of Hematology, Urumqi General Hospital of Chinese People's Liberation Army, Urumqi, Xinjiang 830000, P.R. China
| | - Song Mei
- Burns and Plastic Surgery Center of People's Liberation Army, Lanzhou General Hospital of Chinese People's Liberation Army, Lanzhou, Gansu 730050, P.R. China
| | - Xiao Bin
- Burns and Plastic Surgery Center of People's Liberation Army, Lanzhou General Hospital of Chinese People's Liberation Army, Lanzhou, Gansu 730050, P.R. China
| | - Hu Da-Hai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Liu Yi
- Burns and Plastic Surgery Center of People's Liberation Army, Lanzhou General Hospital of Chinese People's Liberation Army, Lanzhou, Gansu 730050, P.R. China
| |
Collapse
|
43
|
Bian EB, Chen EF, Xu YD, Yang ZH, Tang F, Ma CC, Wang HL, Zhao B. Exosomal lncRNA‑ATB activates astrocytes that promote glioma cell invasion. Int J Oncol 2018; 54:713-721. [PMID: 30483768 DOI: 10.3892/ijo.2018.4644] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/29/2018] [Indexed: 11/05/2022] Open
Abstract
Glioma invasion is a main cause of a poor prognosis and relapse in patients suffering from the disease. However, the molecular mechanisms responsible for glioma cell invasion remain poorly understood. In this study, the characteristics of exosomes were identified using electron microscope (TEM), and western blot analysis. The potential mechanism of long non‑coding RNA (lncRNA) activated by TGF‑β (lncRNA‑ATB) was demonstrated using luciferase reporter assays and RNA immunoprecipitation. We found that glioma cell‑derived exosomes promoted the activation of astrocytes and had the ability to shuttle long non‑coding RNA (lncRNA) activated by TGF‑β (lncRNA‑ATB) to astrocytes. More importantly, lncRNA‑ATB activated astrocytes through the suppression of microRNA (miRNA or miR)‑204‑3p in an Argonaute 2 (Ago2)‑dependent manner. Furthermore, astrocytes activated by lncRNA‑ATB in turn promoted the migration and invasion of glioma cells. Taken together, the findings of this study suggest that lncRNA‑ATB may play an important role in modulating glioma microenvironment through exosomes. Thus, a better understanding of this process may provide implications for the prevention of highly invasive glioma.
Collapse
Affiliation(s)
- Er-Bao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Er-Feng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Ya-Di Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Zhi-Hao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Feng Tang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Chun-Chun Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Hong-Liang Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
44
|
Jin H, Jin X, Chai W, Yin Z, Li Y, Dong F, Wang W. Long non-coding RNA MIAT competitively binds miR-150-5p to regulate ZEB1 expression in osteosarcoma. Oncol Lett 2018; 17:1229-1236. [PMID: 30655889 DOI: 10.3892/ol.2018.9671] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 06/19/2018] [Indexed: 11/05/2022] Open
Abstract
Long non-coding RNAs (LncRNAs), are significant in a number of biological stages and illnesses. The myocardial infarction associated transcript (MIAT) serves a function in numerous types of illness and physiological and pathological processes, including paranoid schizophrenia, diabetic retinopathy, myocardial infarction and neuroendocrine prostate cancer. However, the function of the lncRNA MIAT in the development of osteosarcoma is unknown. It has been identified that during the development of osteosarcoma, MIAT is upregulated in tumor tissues compared to adjacent non-tumor tissues. The spreading and proliferation of osteosarcoma cells was reduced by MIAT knockdown. These findings indicate that MIAT functions by competing with critical RNAs to target miR-150-5p and activate zinc finger E-box binding homeobox 1 to modulate the function of osteosarcoma cells. Together, the present findings may contribute to the understanding of the pathogenesis of osteosarcoma.
Collapse
Affiliation(s)
- Hao Jin
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Xin Jin
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Weiguang Chai
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Zhiqiang Yin
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Yang Li
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Feng Dong
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| | - Wenbo Wang
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150006, P.R. China
| |
Collapse
|
45
|
Jiang X, Zhang C, Qi S, Guo S, Chen Y, Du E, Zhang H, Wang X, Liu R, Qiao B, Yang K, Zhang Z, Xu Y. Elevated expression of ZNF217 promotes prostate cancer growth by restraining ferroportin-conducted iron egress. Oncotarget 2018; 7:84893-84906. [PMID: 27768596 PMCID: PMC5356707 DOI: 10.18632/oncotarget.12753] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/05/2016] [Indexed: 01/09/2023] Open
Abstract
Although we and other studies indicated ZNF217 expression was increased in prostate cancer (PCa), the factors mediating its misregulated expression and their oncogenic activity remain largely unexplored. Recent evidence demonstrated that ferroportin (FPN) reduction lead to decreased iron export and increased intercellular iron that consequently aggravates the oncogenic effects of iron. In the present study, ZNF217 was identified as a transcriptional repressor that inhibits FPN expression. Increased of ZNF217 expression led to decreased FPN concentration, coupled with resultant intracellular iron retention, increased iron-related cellular activities and enhanced tumor cell growth. In contrast, decreased of ZNF217 expression restrained tumor cell growth by promoting FPN-driven iron egress. Mechanistic investigation manifested that ZNF217 facilitated the H3K27me3 levels of FPN promoter by interacting with EZH2. Besides, we also found that MAZ increased the transcription level of ZNF217, and subsequently inhibited the FPN expression and their iron–related activities. Strikingly, the expression of MAZ, EZH2 and ZNF217 were concurrently upregulated in PCa, leading to decreased expression of FPN, which induce disordered iron metabolism. Collectively, this study underscored that elevated expression of ZNF217 promotes prostate cancer growth by restraining FPN-conducted iron egress.
Collapse
Affiliation(s)
- Xingkang Jiang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Changwen Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Shiyong Qi
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Shanqi Guo
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300112, China
| | - Yue Chen
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - E Du
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Hongtuan Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Xiaoming Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Ranlu Liu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Baomin Qiao
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Kuo Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Zhihong Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Yong Xu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| |
Collapse
|
46
|
Liu J, Zhu H, Wang H, Li J, Han F, Liu Y, Zhang W, He T, Li N, Zheng Z, Hu D. Methylation of secreted frizzled-related protein 1 (SFRP1) promoter downregulates Wnt/β-catenin activity in keloids. J Mol Histol 2018; 49:185-193. [DOI: 10.1007/s10735-018-9758-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/05/2018] [Indexed: 01/13/2023]
|
47
|
Zheng W, Yu A. EZH2-mediated suppression of lncRNA-LET promotes cell apoptosis and inhibits the proliferation of post-burn skin fibroblasts. Int J Mol Med 2018; 41:1949-1957. [PMID: 29393360 PMCID: PMC5810232 DOI: 10.3892/ijmm.2018.3425] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/09/2017] [Indexed: 12/28/2022] Open
Abstract
Although the upregulation of enhancer of zeste homolog 2 (EZH2) expression and downregulation of long non-coding RNA (lncRNA) LET expression are known to be associated with cell apoptosis and proliferation, little is known about the interaction of EZH2 with lncRNA LET. The present study aimed to investigate the interaction of EZH2 and lncRNA LET, and the mechanism of human dermal fibroblast (HDF) proliferation and apoptosis. Tissue samples from 33 burn patients with second- and third-degree burns and 8 controls were collected. mRNA was extracted from the burn tissues for analysis. Isolated primary HDFs were treated with heat or transfected with LET overexpression vectors, and the cell functions and associated proteins in the HDFs were analyzed. Decreased lncRNA LET expression was detected in burn tissues compared with normal skin. Heat-treated HDFs exhibited a reduction in lncRNA LET expression and increase in EZH2 expression. LET gain-of-function experiments in primary HDFs revealed increases in cell proliferation, the proportion of cells in the S stage, and cyclin D1 and cyclin-dependent kinase 4 (CDK4) expression, and reductions in the percentage of apoptotic cells, the Bax/Bcl-2 ratio and caspase-3 expression. RNA immunoprecipitation and chromatin immunoprecipitation assays demonstrated the interaction of ZH2 with lncRNA LET, and of EZH2 with H3K27me3 in HDFs. Furthermore, a negative correlation between lncRNA LET and EZH2 expression was identified. It may be concluded that increased lncRNA-LET expression promoted cell proliferation and inhibited cell apoptosis via the cyclin D1-CDK4 and Bax/Bcl-2/caspase-3 signaling pathways, respectively. Furthermore, the inhibition of lncRNA LET may be regarded as an option for use in the healing of burns.
Collapse
Affiliation(s)
- Weicai Zheng
- Department of Burns and Plastic Surgery, The First Affiliated Hospital of Henan University of Science and Technology, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Aixiang Yu
- Department of Burns and Plastic Surgery, The First Affiliated Hospital of Henan University of Science and Technology, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
48
|
Long non-coding RNA expression profiling in the lesional tissue and derived fibroblasts of keloid. Postepy Dermatol Alergol 2017; 34:587-600. [PMID: 29422825 PMCID: PMC5799763 DOI: 10.5114/ada.2017.72466] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
Introduction Long non-coding RNA (lncRNA) plays a key role in various disorders. However, its role in keloid is still unclear. Aim We explored differentially expressed (DE) lncRNAs and mRNAs between keloid tissue (KT)s and normal tissue (NT)s, as well as keloid fibroblast (KFB)s and normal fibroblast (NFB)s, respectively. Material and methods We use KTs and NTs from the chest of 5 patients, and 3 pairs of KFBs and NFBs, to perform microarray respectively. Gene ontology and pathway analyses were conducted by online software DAVID (Database for Annotation, Visualization and Integrated Discovery). The validation of targeted lncRNAs were conducted by qRT-PCR in enlarged samples (79 KTs and 21 NTs). Results We identified 3680 DE-lncRNAs in tissue essay, and 1231 DE-lncRNAs in cell essay. Furthermore, we found that many lncRNAs and their relative mRNAs were regulated simultaneously in keloid. We identified that ENST00000439703 and uc003jox.1 were up-regulated in both of the above essays through comparing the results of lncRNA screening between tissue essay and cell essay; the results were confirmed through qRT-PCR in enlarged samples. Conclusions Our study demonstrates that numerous lncRNAs are involved in the pathogenesis and development of the keloid.
Collapse
|
49
|
Malhotra A, Jain M, Prakash H, Vasquez KM, Jain A. The regulatory roles of long non-coding RNAs in the development of chemoresistance in breast cancer. Oncotarget 2017; 8:110671-110684. [PMID: 29299178 PMCID: PMC5746413 DOI: 10.18632/oncotarget.22577] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/13/2017] [Indexed: 01/01/2023] Open
Abstract
Chemoresistance is one of the major hurdles in the treatment of breast cancer, which limits the effect of both targeted and conventional therapies in clinical settings. Therefore, understanding the mechanisms underpinning resistance is paramount for developing strategies to circumvent resistance in breast cancer patients. Several published reports have indicated that lncRNAs play a dynamic role in the regulation of both intrinsic and acquired chemoresistance through a variety of mechanisms that endow cells with a drug-resistant phenotype. Although a number of lncRNAs have been implicated in chemoresistance of breast cancer, their mechanistic roles have not been systematically reviewed. Thus, here we present a detailed review on the latest research findings and discoveries on the mechanisms of acquisition of chemoresistance in breast cancer related to lncRNAs, and how lncRNAs take part in various cancer signalling pathways involved in breast cancer cells. Knowledge obtained from this review could assist in the development of new strategies to avoid or reverse drug resistance in breast cancer chemotherapy.
Collapse
Affiliation(s)
- Akshay Malhotra
- Center for Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Manju Jain
- Center for Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Hridayesh Prakash
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, India
| | - Karen M Vasquez
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, TX, USA
| | - Aklank Jain
- Center for Animal Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
50
|
Liu Y, Li Y, Xu Q, Yao W, Wu Q, Yuan J, Yan W, Xu T, Ji X, Ni C. Long non-coding RNA-ATB promotes EMT during silica-induced pulmonary fibrosis by competitively binding miR-200c. Biochim Biophys Acta Mol Basis Dis 2017; 1864:420-431. [PMID: 29113749 DOI: 10.1016/j.bbadis.2017.11.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/28/2017] [Accepted: 11/02/2017] [Indexed: 10/18/2022]
Abstract
Long non-coding RNAs (lncRNAs) are important signal transduction regulators that act by various patterns. However, little is known about the molecular mechanisms of lncRNA related pathways in occupational lung fibrosis. Our previous study found that epithelial-mesenchymal transition (EMT) was one of the key events in silica-induced pulmonary fibrosis. This study showed that the lncRNA-ATB promoted EMT by acting as a miR-200c sponge. miR-200c was identified by miRNA array as a potential target of lncRNA-ATB and verified by dual luciferase reporter gene together with RNA pull-down assays. Moreover, our findings demonstrated that lncRNA-ATB is abundantly expressed during EMT of lung epithelial cells, which contributes to decreased levels of miR-200c. miR-200c targeted ZEB1 to relief silicosis by blocking EMT in vivo and in vitro. The results also suggested M2 macrophages secreted transforming growth factor-β1 (TGF-β1) to induce EMT process by activating lncRNA-ATB in epithelial cells. Collectively, silica-stimulated macrophages secreted TGF-β1 to induce lncRNA-ATB in epithelia cells, promoting EMT by binding with miR-200c and releasing ZEB1. These observations provide further understanding of the regulatory network of silica-induced pulmonary fibrosis and identify new therapeutic targets hopefully.
Collapse
Affiliation(s)
- Yi Liu
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yan Li
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qi Xu
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenxi Yao
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiuyun Wu
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jiali Yuan
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Weiwen Yan
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Tiantian Xu
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoming Ji
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chunhui Ni
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|