1
|
Taghizadieh M, Kalantari M, Bakhshali R, Kobravi S, Khalilollah S, Baghi HB, Bayat M, Nahand JS, Akhavan-Sigari R. To be or not to be: navigating the influence of MicroRNAs on cervical cancer cell death. Cancer Cell Int 2025; 25:153. [PMID: 40251577 PMCID: PMC12008905 DOI: 10.1186/s12935-025-03786-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
With all diagnostic and therapeutic advances, such as surgery, radiation- and chemo-therapy, cervical cancer (CC) is still ranked fourth among the most frequent cancers in women globally. New biomarkers and therapeutic targets are warranted to be discovered for the early detection, treatment, and prognosis of CC. As component of the non-coding RNA's family, microRNAs (miRNAs) participate in several cellular functions such as cell proliferation, gene expression, many signaling cascades, apoptosis, angiogenesis, etc. MiRNAs can suppress or induce programmed cell death (PCD) pathways by altering their regulatory genes. Besides, abnormal expression of miRNAs weakens or promotes various signaling pathways associated with PCD, resulting in the development of human diseases such as CC. For that reason, understanding the effects that miRNAs exert on the various modes of tumor PCD, and evaluating the potential of miRNAs to serve as targets for induction of cell death and reappearance of chemotherapy. The current study aims to define the effect that miRNAs exert on cell apoptosis, autophagy, pyroptosis, ferroptosis, and anoikis in cervical cancer to investigate possible targets for cervical cancer therapy. Manipulating the PCD pathways by miRNAs could be considered a primary therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Mohammad Taghizadieh
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kalantari
- Department of Biology, Tehran University of health Sciences, Tehran, Iran
| | | | - Sepehr Kobravi
- Department of Oral & Maxillofacial Surgery, Faculty of Dentistry, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
2
|
Lin J, Zhang W, Wang S, Wang C, Zhang R, Yang Y, Zhou C, Zhang L, Tang P, Liu J, Jin X, Ma Y. Astragalin inhibits neuronal excitability and activates neuronal autophagy in the ACC and LH of CFA mice to alleviate inflammatory pain and pain-related emotions. Int Immunopharmacol 2025; 148:114115. [PMID: 39842140 DOI: 10.1016/j.intimp.2025.114115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Astragalin (AST), a natural flavonoid, exhibits anti-inflammatory, anti-cancer, and antioxidant properties. However, its effects and molecular mechanisms in inflammatory pain remain unclear. Therefore, this study aims to investigate the impact of AST on a Complete Freund's Adjuvant (CFA)-induced inflammatory pain mouse model and to elucidate its potential mechanisms. We employed behavioral tests, including the paw withdrawal test (PWT) and open field test (OFT), to assess pain thresholds and emotional changes in mice, while ELISA was utilized to measure the expression of inflammatory factors. Western blot analysis was performed to evaluate the expression of autophagy-related proteins, c-Fos, and pathway-related proteins. Additionally, immunofluorescence staining was conducted to assess the co-localization of neurons with autophagy-related factors and c-Fos. Our findings indicate that AST significantly reduces pain sensitivity and anxiety-like behaviors in CFA mice, similar to the analgesic Naproxen (NAP). AST treatment inhibited the expression of c-Fos, a neuronal excitability marker, in the ACC and LH of CFA mice, while upregulating the expression of autophagy-related proteins. Furthermore,AST modulates the expression of proteins associated with the CXCR4-Beclin1/VPS34 signaling pathway. In conclusion, these results suggest that AST inhibits neuronal excitability and enhances autophagy by modulating the CXCR4-Beclin1/VPS34 signaling pathway in the ACC and LH of CFA mice, leading to a reduction in pain sensitivity and anxiety-like behaviors, thereby producing analgesic effects. This study reveals a novel mechanism for the potential use of AST in the treatment of inflammatory pain and pain-related emotions, offering a promising strategy for clinical applications.
Collapse
Affiliation(s)
- Jiahong Lin
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weishan Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuhan Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Can Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Runheng Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yaqi Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chang Zhou
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Pei Tang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing Liu
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuxin Ma
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
3
|
Chu T, Tong J, Zhu Z, Sun L, Cui J, Jiang Y, Liu J, Ahmad N, Zhang L, Song Y. Estrogen promotes autophagy in the mammary epithelial cells of dairy sheep via the CXCL12/CXCR4 axis. J Anim Sci 2025; 103:skaf064. [PMID: 40341494 PMCID: PMC12062523 DOI: 10.1093/jas/skaf064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/05/2025] [Indexed: 05/10/2025] Open
Abstract
Mammary gland development and lactation in dairy sheep are regulated by hormones and autophagy; however, the role of estrogen-mediated autophagy remains unclear. This study determined that estrogen enhances autophagy, promotes CXCR4 and CXCL12 gene expression, and increases the number of autolysosomes in sheep mammary epithelial cells. Co-treatment with a CXCR4 overexpression vector and the small-molecule alternative of CXCL12, NUCC-390, significantly upregulated ATG5 and LC3 gene expression, increased the abundance of the autophagy-associated protein ATG5 and the LC3II/I ratio, and increased the consumption of the autophagy substrate P62. These results suggest that CXCR4 and CXCL12 signaling promotes autophagy in mammary epithelial cells. Conversely, co-treatment with a CXCR4-specific blocker and estrogen inhibited autophagic changes in ATG5, P62, and LC3 levels, reducing the number of autophagosomes and autolysosomes. Overall, this study demonstrated that estrogen promotes autophagy in sheep mammary epithelial cells through the CXCL12/CXCR4 signaling axis, revealing the underlying mechanisms behind estrogen-mediated autophagy.
Collapse
Affiliation(s)
- Tingting Chu
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Jiashun Tong
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Zhongshi Zhu
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Lei Sun
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Jiuzeng Cui
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Yue Jiang
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Jiaxin Liu
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Naseer Ahmad
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, YangLing District, XianYang City, Shaanxi Province, P.R. China
| |
Collapse
|
4
|
Zhang Q, Song J, Sun M, Xu T, Li S, Fu X, Yin R. RNF113A as a poor prognostic factor promotes metastasis and invasion of cervical cancer through miR197/PRP19/P38MAPK signaling pathway. Arch Biochem Biophys 2024; 761:110139. [PMID: 39242014 DOI: 10.1016/j.abb.2024.110139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/09/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
It has been discovered that aberrant expression of RNF113A plays a significant role in various diseases, including esophageal cancer, hepatocellular carcinoma, and X-linked trichothiodystrophy syndrome. Nevertheless, its functional implications in cervical cancer (CC) remain unclear. The objective of this study was to investigate the role of RNF113A in both the development and prognosis of CC. To achieve this objective, a total of sixty cases were included in the follow-up investigation. The findings revealed a significant up-regulation of RNF113A protein in CC tissues compared to paired paracancerous tissues, and a high expression level of RNF113A was strongly associated with malignant phenotypes such as lymph node metastasis, differentiation degree, depth of invasion, and FIGO stage. Meanwhile, RNF113A was found to be an independent prognostic risk factor, with its high expression significantly correlating with a reduced overall survival period in patients. To elucidate the underlying cause and mechanism of the unfavorable prognosis associated with RNF113A, comprehensive functional investigations were conducted both in vitro and in vivo.Interestingly, it was revealed that RNF113A promoted migration and invasion while inhibiting apoptosis of CC cells, thereby contributing to a poor prognosis. Mechanistically, RNF113A regulated the progression and prognosis of CC through the miR197/Prp19/p38Mark signaling pathway. Overall, our findings underscore the potential clinical significance of RNF113A as an unfavorable prognostic factor in CC.
Collapse
Affiliation(s)
- Qingwei Zhang
- The Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, 610041, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China
| | - Jiayu Song
- Department of Pharmacology, Luohe Medical College, Luohe, 462000, Henan, China
| | - Mingzhen Sun
- Department of Pharmacology, Luohe Medical College, Luohe, 462000, Henan, China
| | - Tenghan Xu
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China; Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China
| | - Suhong Li
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China; Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China
| | - Xiuhong Fu
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe, 462000, China; Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe, 462000, Henan, China
| | - Rutie Yin
- The Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China.
| |
Collapse
|
5
|
Wen W, Ertas YN, Erdem A, Zhang Y. Dysregulation of autophagy in gastric carcinoma: Pathways to tumor progression and resistance to therapy. Cancer Lett 2024; 591:216857. [PMID: 38583648 DOI: 10.1016/j.canlet.2024.216857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
The considerable death rates and lack of symptoms in early stages of gastric cancer (GC) make it a major health problem worldwide. One of the most prominent risk factors is infection with Helicobacter pylori. Many biological processes, including those linked with cell death, are disrupted in GC. The cellular "self-digestion" mechanism necessary for regular balance maintenance, autophagy, is at the center of this disturbance. Misregulation of autophagy, however, plays a role in the development of GC. In this review, we will examine how autophagy interacts with other cell death processes, such as apoptosis and ferroptosis, and how it affects the progression of GC. In addition to wonderful its role in the epithelial-mesenchymal transition, it is engaged in GC metastasis. The role of autophagy in GC in promoting drug resistance stands out. There is growing interest in modulating autophagy for GC treatment, with research focusing on natural compounds, small-molecule inhibitors, and nanoparticles. These approaches could lead to breakthroughs in GC therapy, offering new hope in the fight against this challenging disease.
Collapse
Affiliation(s)
- Wen Wen
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey.
| | - Ahmet Erdem
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI, 48824, USA; Department of Biomedical Engineering, Kocaeli University, Umuttepe Campus, Kocaeli, 41001 Turkey.
| | - Yao Zhang
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
6
|
Kobayashi H, Imanaka S, Yoshimoto C, Matsubara S, Shigetomi H. Molecular mechanism of autophagy and apoptosis in endometriosis: Current understanding and future research directions. Reprod Med Biol 2024; 23:e12577. [PMID: 38645639 PMCID: PMC11031673 DOI: 10.1002/rmb2.12577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/29/2024] [Accepted: 04/05/2024] [Indexed: 04/23/2024] Open
Abstract
Background Endometriosis is a common gynecological condition, with symptoms including pain and infertility. Regurgitated endometrial cells into the peritoneal cavity encounter hypoxia and nutrient starvation. Endometriotic cells have evolved various adaptive mechanisms to survive in this inevitable condition. These adaptations include escape from apoptosis. Autophagy, a self-degradation system, controls apoptosis during stress conditions. However, to date, the mechanisms regulating the interplay between autophagy and apoptosis are still poorly understood. In this review, we summarize the current understanding of the molecular characteristics of autophagy in endometriosis and discuss future therapeutic challenges. Methods A search of PubMed and Google Scholar databases were used to identify relevant studies for this narrative literature review. Results Autophagy may be dynamically regulated through various intrinsic (e.g., PI3K/AKT/mTOR signal transduction network) and extrinsic (e.g., hypoxia and iron-mediated oxidative stress) pathways, contributing to the development and progression of endometriosis. Upregulation of mTOR expression suppresses apoptosis via inhibiting the autophagy pathway, whereas hypoxia or excess iron often inhibits apoptosis via promoting autophagy. Conclusion Endometriotic cells may have acquired antiapoptotic mechanisms through unique intrinsic and extrinsic autophagy pathways to survive in changing environments.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive MedicineMs.Clinic MayOneKashiharaJapan
- Department of Obstetrics and GynecologyNara Medical UniversityKashiharaJapan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive MedicineMs.Clinic MayOneKashiharaJapan
- Department of Obstetrics and GynecologyNara Medical UniversityKashiharaJapan
| | - Chiharu Yoshimoto
- Department of Obstetrics and GynecologyNara Medical UniversityKashiharaJapan
- Department of Obstetrics and GynecologyNara Prefecture General Medical CenterNaraJapan
| | - Sho Matsubara
- Department of Obstetrics and GynecologyNara Medical UniversityKashiharaJapan
- Department of MedicineKei Oushin ClinicNishinomiyaJapan
| | - Hiroshi Shigetomi
- Department of Obstetrics and GynecologyNara Medical UniversityKashiharaJapan
- Department of Gynecology and Reproductive MedicineAska Ladies ClinicNaraJapan
| |
Collapse
|
7
|
Yu H, Xie L, Chen Z, Niu H, Jia X, Du B, Shen Y, Gui L, Xu X, Li J. miR-22a targets p62/SQSTM1 to negatively affect autophagy and disease resistance of grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2023; 142:109124. [PMID: 37777097 DOI: 10.1016/j.fsi.2023.109124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
MicroRNAs (miRNAs) are integral to many biological functions, including autophagy, a process recently proven to be closely linked to innate immunity. In this study, we present findings on miR-22a, a teleost homolog of mammalian miR-22, illustrating its capacity to target the autophagy adaptor p62, subsequently inducing downregulation at both mRNA and protein levels. Utilizing Western blot analyses, we demonstrated that miR-22a inhibits the autophagy flux of CIK cells, correlated with an elevated presence of LC3 II. Additionally, the overexpression of miR-22a resulted in the suppression of NF-κB signaling, leading to reduced cellar antimicrobial abilities and increased apoptosis. These findings provide novel insights into the role of miR-22a as an autophagy-related miRNA and its immune mechanisms against pathogens via p62 in teleost, enriching our understanding of the interplay between autophagy and innate immunity.
Collapse
Affiliation(s)
- Hongyan Yu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Lingli Xie
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Zheyan Chen
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Huiqin Niu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Xuewen Jia
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Biao Du
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yubang Shen
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Lang Gui
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xiaoyan Xu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| | - Jiale Li
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|