1
|
Manjili DA, Babaei FN, Younesirad T, Ghadir S, Askari H, Daraei A. Dysregulated circular RNA and long non-coding RNA-Mediated regulatory competing endogenous RNA networks (ceRNETs) in ovarian and cervical cancers: A non-coding RNA-Mediated mechanism of chemotherapeutic resistance with new emerging clinical capacities. Arch Biochem Biophys 2025; 768:110389. [PMID: 40090441 DOI: 10.1016/j.abb.2025.110389] [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: 11/11/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Cervical cancer (CC) and ovarian cancer (OC) are among the most common gynecological cancers with significant mortality in women, and their incidence is increasing. In addition to the prominent role of the malignant aspect of these cancers in cancer-related women deaths, chemotherapy drug resistance is a major factor that contributes to their mortality and presents a clinical obstacle. Although the exact mechanisms behind the chemoresistance in these cancers has not been revealed, accumulating evidence points to the dysregulation of non-coding RNAs (ncRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as key contributors. These ncRNAs perform the roles of regulators of signaling pathways linked to tumor formation and chemoresistance. Strong data from various recent studies have uncovered that the main mechanism of these ncRNAs in the induction of chemoresistance of CC and OC is done through a dysregulated miRNA sponge activity as competing endogenous RNA (ceRNA) in the competing endogenous RNA networks (ceRNETs), where a miRNA regulating a messenger RNA (mRNA) is trapped, thereby removing its inhibitory effect on the desired mRNA. Understanding these mechanisms is essential to enhancing treatment outcomes and managing the problem of drug resistance. This review provides a comprehensive overview of lncRNA- and circRNA-mediated ceRNETs as the core process of chemoresistance against the commonly used chemotherapeutics, including cisplatin, paclitaxel, oxaliplatin, carboplatin, and docetaxel in CC and OC. Furthermore, we highlight the clinical potential of these ncRNAs serving as diagnostic indicators of chemotherapy responses and therapeutic targets.
Collapse
Affiliation(s)
- Danial Amiri Manjili
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Naghdi Babaei
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tayebeh Younesirad
- Department of Medical Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sara Ghadir
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Askari
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
2
|
Liu Y, Wang Y, Yan P, Cui N, Xu K, Liu D, Tian Y, Cao L. NLRP3 Inflammasome-Mediated Osteoarthritis: The Role of Epigenetics. BIOLOGY 2025; 14:71. [PMID: 39857301 PMCID: PMC11761621 DOI: 10.3390/biology14010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
The prevalence of osteoarthritis (OA) notably surges with age and weight gain. The most common clinical therapeutic drugs are painkillers, yet they cannot impede the deteriorating course of OA. Thus, understanding OA's pathogenesis and devising effective therapies is crucial. It is generally recognized that inflammation, pyroptosis, and OA progression are tightly linked. The activation of NLRP3 inflammasome can lead to the discharge of the pro-inflammatory cytokines Interleukin-1β and IL-18, intensifying subsequent inflammatory reactions and promoting OA development. Conversely, the imbalance caused by deacetylase-regulated NLRP3 inflammasome underlies the chronic mild inflammation related to degenerative diseases. Therefore, this article expounds on the mechanism of OA pathogenesis and the role of histone deacetylases (HDACs) in NLRP3 inflammasome-triggered OA, and illustrates the application of HDAC inhibitors in OA, striving to provide more insights into novel OA treatment approaches.
Collapse
Affiliation(s)
- Yuzhou Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (Y.L.); (Y.W.); (K.X.)
| | - Ying Wang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (Y.L.); (Y.W.); (K.X.)
| | - Ping Yan
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (P.Y.); (N.C.)
| | - Ning Cui
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (P.Y.); (N.C.)
| | - Kejin Xu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (Y.L.); (Y.W.); (K.X.)
| | - Da Liu
- Public Laboratory Centre, Changchun University of Chinese Medicine, Changchun 130117, China;
| | - Yuan Tian
- Clinical School of Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China
| | - Lingling Cao
- Clinical School of Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China
| |
Collapse
|
3
|
Ellakwa DES, Abdelmalek MA, Mostafa MM, Ellakwa TE, Wadan AHS. MircoRNAs predict and modulate responses to chemotherapy in leukemic patients. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03675-7. [PMID: 39808312 DOI: 10.1007/s00210-024-03675-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/24/2024] [Indexed: 01/16/2025]
Abstract
Leukemia covers a broad category of cancer malignancies that specifically affect bone marrow and blood cells. While different kinds of leukemia have been identified, effective treatments are still lacking for most forms, and even those treatments considered effective can lead to relapses. MicroRNAs, or miRNAs, are short endogenous non-coding single-stranded RNAs that help control the epigenetics of gene expression. Recently, a literature review proposed that miRNAs provided promising therapeutic targets for patients diagnosed with leukemia. Due to genetic abnormalities occurring during the maturation of white blood cells, studies commonly observed uncontrolled replication and decreased cell death, compared to healthy cells. This results in the activation of oncogenes, deactivation of tumor suppressor genes, and disruption of normal cellular functions. Although conventional cancer treatments significantly contribute to patient recovery, they can also impose many side effects. MiRNAs all significantly regulate angiogenesis, migration, apoptosis, carcinogenesis, and gene expression. Regarding chemotherapy, mounting research indicates that microRNAs may directly influence how responsive leukemia is to chemical treatments. This article reviews current studies on microRNAs, examining their influence on cancer advancement and spread, as well as their possible applications as diagnostic indicators and treatment targets in leukemia. Furthermore, we integrated the functions of microRNAs in cancer formation and progression with leukemia patient care, offering fresh insights into leukemia detection and management strategies.
Collapse
Affiliation(s)
- Doha El-Sayed Ellakwa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo, Egypt.
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt.
| | | | - Mostafa M Mostafa
- Department of Molecular and Cellular Physiology, Stritch School of Medicine, Loyola University Chicago, Chicago, USA
| | - Takwa E Ellakwa
- Physical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Al-Hassan Soliman Wadan
- Oral Biology Department, Faculty of Dentistry, Galala Plateau, Galala University, 15888), Attaka, Suez Governorate, Egypt
| |
Collapse
|
4
|
Pan H, Zhang W, Qin Z, Jia K, Jia P, Yi M. MiR-192 and miR-731 synergically inhibit RGNNV infection by targeting ULK1-mediated autophagy in sea perch (Lateolabrax japonicus). Int J Biol Macromol 2024; 282:136748. [PMID: 39437957 DOI: 10.1016/j.ijbiomac.2024.136748] [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: 07/15/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
MicroRNAs (miRNAs) are crucial regulators of gene expression and are closely linked to viral infections. Nervous necrosis virus (NNV) poses a significant threat to global fish aquaculture. This study investigates the roles of miR-192 and miR-731 in controlling NNV infection and associated autophagy in sea perch (Lateolabrax japonicus). Our findings reveal that both miRNAs are upregulated in infected brain tissues and cells of sea perch, leading to reduced NNV replication. miR-192 and miR-731 synergistically reduce NNV replication by downregulating the RNA-dependent RNA polymerase and capsid protein genes, and disrupting autophagy by altering LC3 distribution and autophagy-related protein expressions. Additionally, L. japonicus unc-51 like autophagy activating kinase 1 protein (LjULK1) was identified as the target of miR-192 and miR-731. LjULK1 is integrally associated to the ULK-VAPs-Atg13 autophagic signaling, enhancing NNV-induced autophagy, and facilitating viral infection. Modulating LjULK1 expression counteracts the inhibitory effects of miR-192 and miR-731 on NNV, suggesting these miRNAs act as negative regulators of NNV infection by targeting LjULK1-mediated autophagy. Our findings reveal a novel miRNAs-regulated antiviral mechanism against NNV, offering insights into potential strategy to prevent NNV infection in fish.
Collapse
Affiliation(s)
- Hongbo Pan
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Zhuhai, Guangdong 519082, China
| | - Wanwan Zhang
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Zhuhai, Guangdong 519082, China
| | - Ziling Qin
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Zhuhai, Guangdong 519082, China
| | - Kuntong Jia
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Zhuhai, Guangdong 519082, China; State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Peng Jia
- Fuzhou Medical College of Nanchang University, Fuzhou 344100, China.
| | - Meisheng Yi
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Zhuhai, Guangdong 519082, China.
| |
Collapse
|
5
|
Wang Y, Kong R, Xie K, Wang X, Wu H, Zhang Y. The Chicken HDAC4 Promoter and Its Regulation by MYC and HIF1A. Genes (Basel) 2024; 15:1518. [PMID: 39766786 PMCID: PMC11675110 DOI: 10.3390/genes15121518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Histone deacetylase 4 (HDAC4) is a member of the class II histone deacetylase family, whose members play a crucial role in various biological processes. An in-depth investigation of the transcriptional characteristics of chicken HDAC4 can provide fundamental insights into its function. METHODS We examined HDAC4 expression in chicken embryonic stem cells (ESC) and spermatogonial stem cells (SSC) and cloned a 444 bp fragment from upstream of the chicken HDAC4 transcription start site. Subsequently, we constructed pEGFP-HDAC4 and a series of 5'-deletion luciferase reporter constructs, which we transfected into DF-1 cells to measure their transcriptional activity. The regulatory mechanisms of chicken HDAC4 expression were investigated by performing trichostatin A (TSA) treatment, deleting putative transcription factor binding sites, and altering transcription factor expression levels. RESULTS HDAC4 exhibited higher expression in SSC than in ESC. We confirmed that the upstream region from -295 bp to 0 bp is the core transcriptional region of HDAC4. TSA effectively inhibited HDAC4 transcription, and bioinformatics analysis indicated that the chicken core HDAC4 promoter sequence exhibits high homology with those of other avian species. The myelocytomatosis viral oncogene homolog (MYC) and hypoxia-inducible factor 1 α (HIF1A) transcription factors were predicted to bind to this core region. Treatment with TSA for 24 h resulted in the upregulation of MYC and HIF1A, which repressed HDAC4 transcription. CONCLUSIONS Our results provide a basis for subsequent investigations into the regulation of HDAC4 expression and biological function.
Collapse
Affiliation(s)
- Yingjie Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Ruihong Kong
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Ke Xie
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Xu Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Han Wu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yani Zhang
- Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
6
|
Lei M, Liang J, Guo K, Tang L, He Y, Wu X. Roles of noncoding RNAs in multiple myeloma. Leuk Res 2024; 146:107593. [PMID: 39307099 DOI: 10.1016/j.leukres.2024.107593] [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: 06/03/2024] [Accepted: 09/16/2024] [Indexed: 11/05/2024]
Abstract
Noncoding RNAs (ncRNAs) constitute a class of nucleic acid molecules within cells that do not encode proteins but play important roles in regulating gene expression, maintaining cellular homeostasis, and mediating cell signaling. This class encompasses microRNAs (miRNAs), long noncoding RNAs (lncRNAs), transfer RNAs (tRNAs), circular RNAs (circRNAs), small interfering RNAs (siRNAs), and others. miRNAs are pivotal in the regulation of gene expression in hematologic malignancies. Aberrant expression of lncRNAs has been confirmed in cancerous tissues, implicating their involvement in carcinogenesis or tumor suppression processes. tRNAs may induce errors or disturbances in protein synthesis, thereby affecting normal cellular function and proliferation. Moreover, circRNAs influence disease progression in tumors by modulating the expression of relevant genes, and siRNAs can inhibit tumor cell proliferation, invasion, and metastasis while inducing apoptosis. This review will elucidate the biological functions of ncRNAs in multiple myeloma (MM) and explore their potential value as therapeutic targets.
Collapse
Affiliation(s)
- Ming Lei
- Changde Hospital, Xiangya School of Medicine, Central South University(The First People's Hospital of Changde City), China
| | - Juan Liang
- Hengyang Medical College, University of South China, China
| | - Kaiyun Guo
- Changde Hospital, Xiangya School of Medicine, Central South University(The First People's Hospital of Changde City), China
| | - Langui Tang
- Changde Hospital, Xiangya School of Medicine, Central South University(The First People's Hospital of Changde City), China
| | - Yuxing He
- Changde Hospital, Xiangya School of Medicine, Central South University(The First People's Hospital of Changde City), China
| | - Xuefeng Wu
- Changde Hospital, Xiangya School of Medicine, Central South University(The First People's Hospital of Changde City), China.
| |
Collapse
|
7
|
Wu Y, Chen Y, Tian X, Shao G, Lin Q, Sun A. Ubiquitination regulates autophagy in cancer: simple modifications, promising targets. J Transl Med 2024; 22:985. [PMID: 39482684 PMCID: PMC11526641 DOI: 10.1186/s12967-024-05565-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/02/2024] [Indexed: 11/03/2024] Open
Abstract
Autophagy is an important lysosomal degradation process that digests and recycles bio-molecules, protein or lipid aggregates, organelles, and invaded pathogens. Autophagy plays crucial roles in regulation of metabolic and oxidative stress and multiple pathological processes. In cancer, the role of autophagy is dual and paradoxical. Ubiquitination has been identified as a key regulator of autophagy that can influence various steps in the autophagic process, with autophagy-related proteins being targeted for ubiquitination, thus impacting cancer progression and the effectiveness of therapeutic interventions. This review will concentrate on mechanisms underlying autophagy, ubiquitination, and their interactions in cancer, as well as explore the use of drugs that target the ubiquitin-proteasome system (UPS) and ubiquitination process in autophagy as part of cancer therapy.
Collapse
Affiliation(s)
- Yihui Wu
- Institute of Urinary System Diseases, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang, 212002, China
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Yifei Chen
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xianyan Tian
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Genbao Shao
- Institute of Urinary System Diseases, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang, 212002, China
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Qiong Lin
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| | - Aiqin Sun
- Institute of Urinary System Diseases, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang, 212002, China.
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
8
|
Sarafraznia L, Tahan Nejad Asadi Z, Dayer D, Jalalifar MA, Ghanatir N. Investigation of Non-Coding RNA-Related Autophagy Alterations in Drug-Resistant Multiple Myeloma Plasma Cells. IRANIAN JOURNAL OF PATHOLOGY 2024; 19:422-430. [PMID: 40034935 PMCID: PMC11872035 DOI: 10.30699/ijp.2024.2022061.3256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/14/2024] [Indexed: 03/05/2025]
Abstract
Background & Objective Multiple myeloma (MM) drug resistance is thought to be caused by the development of protective autophagy. This work aimed to assess the non-coding RNA (ncRNA) autophagy-related alterations in drug-resistant (DR) myeloma cells. Methods DR Plasma cells were extracted from the bone marrow of DR patients referred to Baghai 2 Hospital in Ahvaz, Iran. The cells were grown in RPMI-1640 media containing 10% FBS and 1% Pen/Strep and incubated at 37˚C and 5% CO2. After six passages, the plasma cells were precisely isolated and utilized as DR cells. The U266B1 cell line (IBRC C10148) was grown in the RPMI-1640 media containing 10% FBS and 1% Pen/Strep and utilized as drug-sensitive (DS) cells. The relative expression of the genes was determined using the Real-time PCR method. Statistical analysis of the data was performed using GraphPad Prism 8 software. Results When the DR cells were compared to the DS cells, there was a notable increase in the expression of ULK1 and LC3B. However, expression of P62 in the DR plasma cells showed a significant decrease compared to the DS plasma cells. The miR-1297 level was considerably higher in the DR cells than in the DS cells. Although, there was no statistically significant difference in the expression of miR-26a-5p between the DS and DR cells. The DR cells exhibited a statistically significant increase in the expression of MALAT1 and SNHG6. Conclusion Drug resistance in MM cells may result from overexpression of non-coding RNAs involved in autophagy.
Collapse
Affiliation(s)
- Leila Sarafraznia
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Thalassemia & Hemoglobinopathy Research center, Health research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zari Tahan Nejad Asadi
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Thalassemia & Hemoglobinopathy Research center, Health research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Dian Dayer
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Ali Jalalifar
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Thalassemia & Hemoglobinopathy Research center, Health research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nariman Ghanatir
- Thalassemia & Hemoglobinopathy Research center, Health research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
9
|
Ma Y, Guo Y, Yuan G, Huang T. Triclosan impairs spermatocyte cell proliferation and induces autophagy by regulating microRNA-20a-5 P by pargeting PTEN. Reprod Toxicol 2024; 129:108663. [PMID: 39002938 DOI: 10.1016/j.reprotox.2024.108663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/09/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Triclosan (TCS), as an endocrine disrupter, has been found to affect male fertility. However, the potential molecular mechanism is still unknown. We aimed to investigate whether the toxic effects of TCS on spermatocyte cells was mediated by the regulation of microRNA-20a-5 P on PTEN. METHODS GC-2 and TM4 cells were treated with TCS (0.5-80 μM) for 24 or 48 hours. Effect of TCS on proliferation of GC-2 and TM4 cells was detected using a cell counting kit-8 (CCK8) assay. Expression of miR-17 family and autophagy genes were detected. The interaction between miR-20a-5 P and PTEN was determined by a dual-luciferase reporter assay. RESULTS TCS decreased cell proliferation of GC-2 and TM4 cells. Expression of autophagy-related genes and miR-17 family was altered by TCS. PTEN expression was significantly increased, whereas the expression of miR-20a-5 P was significantly decreased in GC-2 and TM4 cells. As predicted in relevant databases, there is a binding site of miR-20a-5 P in PTEN. The expression of PTEN was significantly down-regulated by the miR-20a-5 P mimic. CONCLUSION As a downstream target of miR-20a-5 P, PTEN functioned in the autophagy process of which TCS inhibited the proliferation of spermatocyte cells. Our results provided new ideas for revealing the molecular mechanism and protective strategy on male infertility.
Collapse
Affiliation(s)
- Yue Ma
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong 518112, China
| | - Yinsheng Guo
- Department of Public Health Emergency Preparedness and Response, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong 518055, China.
| | - Guanxiang Yuan
- Department of Public Health Emergency Preparedness and Response, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong 518055, China
| | - Ting Huang
- Department of Preventive Medicine and Healthcare-Associated Infection Management, National Clinical Research Center for Infectious Diseases, Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong 518112, China.
| |
Collapse
|
10
|
Zou R, Wang Y, Cai Y, Xing Z, Shao Y, Li D, Qi C. Nanofiber-based delivery of evodiamine impedes malignant properties of intrahepatic cholangiocarcinoma cells by targeting HDAC4 and restoring TPM1 transcription. Hum Cell 2024; 37:1505-1521. [PMID: 39073525 DOI: 10.1007/s13577-024-01105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
The electrospun nanofiber system is correlated with high efficacy of drug delivery. This study aims to investigate the effect of nanofiber-based delivery of evodiamine, an indole alkaloid derived from Rutaceae plants Evodia rutaecarpa (Juss.) Benth, on intrahepatic cholangiocarcinoma (ICC), as well as to explore the molecular mechanisms. An electrospun nanofiber system carrying evodiamine was generated. Compared to evodiamine treatment alone, the nano-evodiamine exhibited more pronounced effects on suppressing proliferation, colony formation, invasiveness, migration, apoptosis resistance, cell cycle progression, and in vivo tumorigenesis of two ICC cell lines (HUCC-T1 and RBE). ICC cells exhibited increased expression of histone deacetylase 4 (HDAC4) while decreased tropomyosin 1 (TPM1). HDAC4 suppressed TPM1 expression by removing H3K9ac modifications from its promoter. Nano-evodiamine reduced HDAC4 protein levels in ICC cells, thus promoting transcription and expression of TPM1. Either overexpression of HDAC4 or downregulation of TPM1 negated the tumor-suppressive effects of nano-evodiamine. Collectively, this study demonstrates that the electrospun nanofiber system enhances the efficiency of evodiamine. Additionally, evodiamine suppresses the malignant properties of ICC cells. The findings may provide fresh insights into the application of electrospun nanofiber system for drug delivery and the effects of evodiamine on tumor suppression.
Collapse
Affiliation(s)
- Rui Zou
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China
| | - Yiyao Wang
- Department of Integrated Traditional Chinese and Western Medicine, Hainan Cancer Hospital, Haikou, 570100, Hainan, People's Republic of China
| | - Yaoqing Cai
- Department of Gastrointestinal Surgery, Hainan Cancer Hospital, Haikou, 570100, Hainan, People's Republic of China
| | - Zhenming Xing
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China
| | - Yongfu Shao
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China
| | - Duo Li
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China.
| | - Chunchun Qi
- Medical College of Nankai University, Tianjin, 300071, People's Republic of China.
| |
Collapse
|
11
|
Pan L, Qin Z, Zhou Q, Zheng P, Li H, Zhou X, Qin Y. Unlocking the Therapeutic Potential: Harnessing miR-125a-5p To Enhance Autophagy and Apoptosis in Pancreatic Cancer through Targeting STAT3. J Cancer 2024; 15:4955-4968. [PMID: 39132168 PMCID: PMC11310883 DOI: 10.7150/jca.97102] [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: 04/08/2024] [Accepted: 06/26/2024] [Indexed: 08/13/2024] Open
Abstract
Objectives: miR-125a-5p's role in various cancers has been recognized, yet its specific function in pancreatic cancer (PCa) demands further exploration. This study aimed to reveal the potential function of miR-125a-5p in PCa. Methods: With publicly available databases, we explored the expression pattern and prognostic relevance of miR-125a-5p and STAT3 in PCa. We measured miR-125a-5p levels in PCa tissues, plasma and cell lines using RT-qPCR. To assess functional effects, PANC-1 cells were transfected with miR-125a-5p mimics and inhibitors, as well as siRNA-STAT3 and STAT3 vectors. Cell proliferation was estimated using Cell Counting Kit-8, while autophagy and apoptosis were examined by transmission electron microscopy and TUNEL assay, respectively. Western blot analysis was also performed to detect proteins associated with autophagy and apoptosis. The regulatory relationship of miR-125a-5p on STAT3 was verified using a dual luciferase reporter assay. The influence of miR-125a-5p on tumor development was evaluated in xenograft models. Results: Decreased expression of miR-125a-5p was found in PCa samples, and low expression of miR-125a-5p was associated with a poorer prognosis in PCa patients. Functional assays indicated miR-125a-5p suppressed cell growth while enhancing apoptosis and autophagy in PCa cells. STAT3 represents a specific target of miR-125a-5p, inhibiting STAT3 reversed the inhibitory effect of overexposed miR-125a-5p. Additionally, miR-125a-5p significantly restrained tumor development in mice. Conclusions: miR-125a-5p functions as a tumor suppressor in PCa by targeting STAT3, thereby inducing autophagy and apoptosis. Its regulatory role underscores its potential as a valuable biomarker for PCa diagnosis and therapy, warranting further clinical investigation.
Collapse
Affiliation(s)
- Lujuan Pan
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
- Key Laboratory of Tumor Molecular Pathology of Baise, Baise, Guangxi Province, China
| | - Zongshuai Qin
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Qinghong Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Pin Zheng
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Hua Li
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Xihan Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Yueqiu Qin
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| |
Collapse
|
12
|
Wang X, Feng Y, Wang F, Lin Z, Huang J, Li Q, Luo H, Liu X, Zhai X, Gao Q, Li L, Zhang Y, Wen J, Zhang L, Niu T, Zheng Y. Single-cell sequencing reveals the correlation of aggrephagy signaling and multiple myeloma immune microenvironment composition. J Gene Med 2024; 26:e3712. [PMID: 38949072 DOI: 10.1002/jgm.3712] [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: 04/27/2023] [Revised: 10/07/2023] [Accepted: 06/02/2024] [Indexed: 07/02/2024] Open
Abstract
Aggrephagy, a type of autophagy, degrades the aggregation of misfolded protein in cells. However, the role of aggrephagy in multiple myeloma (MM) has not been fully demonstrated. In this study, we first investigated the correlation between aggrephagy signaling, MM immune microenvironment composition and disease prognosis. Single-cell RNA-seq data, including the expression profiles of 12,187 single cells from seven MM bone marrow (BM) and seven healthy BM samples, were analyzed by non-negative matrix factorization for 44 aggrephagy-related genes. Bulk RNA-seq cohorts from the Gene Expression Omnibus database were used to evaluate the prognostic value of aggrephagy-related immune cell subtypes and predict immune checkpoint blockade immunotherapeutic response in MM. Compared with healthy BM, MM BM exhibited different patterns of aggrephagy-related gene expression. In MM BM, macrophages, CD8+ T cells, B cells and natural killer cells could be grouped into four to nine aggrephagy-related subclusters. The signature of aggrephagy signaling molecule expression in the immune cells correlates with the patient's prognosis. Our investigation provides a novel view of aggrephagy signaling in MM tumor microenvironment cells, which might be a prognostic indicator and potential target for MM treatment.
Collapse
Affiliation(s)
- Xin Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Feng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhimei Lin
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
- Department of Hematology, The Affiliated Hospital of Chengdu University, Chengdu, China
| | - Jingcao Huang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Luo
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Liu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Zhai
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Qianwen Gao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
- School of Life Science, Sichuan University, Chengdu, China
| | - Linfeng Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Wen
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
- Department of Hematology, Mianyang Central Hospital, Mianyang, China
| | - Li Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Chen Q, Lao J. Interference with Histone Deacetylase 4 Regulates c-Jun N-terminal Kinase/Activating Protein-1 Signaling to Ameliorate Sepsis-induced Alveolar Epithelial Cell Injury. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:174-186. [PMID: 39133038 DOI: 10.4103/ejpi.ejpi-d-24-00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/26/2024] [Indexed: 08/13/2024]
Abstract
ABSTRACT Sepsis is a syndrome of systemic inflammatory response resulting from infection, which can lead to severe lung injury. Histone deacetylase 4 (HDAC4) is a key protein known to regulate a wide range of cellular processes. This study was designed to investigate the role of HDAC4 in lipopolysaccharide (LPS)-induced alveolar epithelial cell injury as well as to disclose its potential molecular mechanisms. The alveolar epithelial cell injury model was established by inducing A549 cells with LPS. A549 cell viability was detected by cell counting kit-8 assay and the transfection efficiency of small interfering RNA targeting HDAC4 was appraised utilizing Western blot. The levels of inflammatory cytokines and oxidative stress markers were detected using corresponding assay kits. Dichloro-dihydro-fluorescein diacetate assay was used for the measurement of reactive oxygen species (ROS) content. Flow cytometry, 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzimidazolyl-carbocyanine iodide-1 staining, adenosine triphosphate (ATP) assay kits, and MitoSOX Red assay kits were employed to estimate cell apoptosis, mitochondrial membrane potential, ATP level, and mitochondrial ROS level, respectively. The oxygen consumption rate of A549 cells was evaluated with XF96 extracellular flux analyzer. Western blot was applied for the evaluation of HDAC4, apoptosis- and c-Jun N-terminal kinase (JNK)/activating protein-1 (AP-1) signaling pathway-related proteins. HDAC4 expression was found to be increased in LPS-induced A549 cells and HDAC4 silence inhibited inflammatory damage, repressed oxidative stress, alleviated cell apoptosis, improved mitochondrial function, and blocked JNK/AP-1 signaling in A549 cells stimulated by LPS, which were all reversed by JNK activator anisomycin. Collectively, the interference with HDAC4 could ameliorate LPS-induced alveolar epithelial cell injury, and such protective effect may be potentially mediated through the JNK/AP-1 signaling pathway.
Collapse
Affiliation(s)
- Qunyan Chen
- Department of Emergency Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | | |
Collapse
|
14
|
Zhang Y, Zhou X. Targeting regulated cell death (RCD) in hematological malignancies: Recent advances and therapeutic potential. Biomed Pharmacother 2024; 175:116667. [PMID: 38703504 DOI: 10.1016/j.biopha.2024.116667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024] Open
Abstract
Regulated cell death (RCD) is a form of cell death that can be regulated by numerous biomacromolecules. Accumulating evidence suggests that dysregulated expression and altered localization of related proteins in RCD promote the development of cancer. Targeting subroutines of RCD with pharmacological small-molecule compounds is becoming a promising therapeutic avenue for anti-tumor treatment, especially in hematological malignancies. Herein, we summarize the aberrant mechanisms of apoptosis, necroptosis, pyroptosis, PANoptosis, and ferroptosis in hematological malignancies. In particular, we focus on the relationship between cell death and tumorigenesis, anti-tumor immunotherapy, and drug resistance in hematological malignancies. Furthermore, we discuss the emerging therapeutic strategies targeting different RCD subroutines. This review aims to summarize the significance and potential mechanisms of RCD in hematological malignancies, along with the development and utilization of pertinent therapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| |
Collapse
|
15
|
Katsaraki K, Kontos CK, Ardavanis-Loukeris G, Tzovaras AA, Sideris DC, Scorilas A. Exploring the time-dependent regulatory potential of microRNAs in breast cancer cells treated with proteasome inhibitors. Clin Transl Oncol 2024; 26:1256-1267. [PMID: 38038871 PMCID: PMC11026233 DOI: 10.1007/s12094-023-03349-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023]
Abstract
PURPOSE Breast cancer (BrCa) is a predominant type of cancer with a disparate molecular nature. MicroRNAs (miRNAs) have emerged as promising key players in the regulation of pathological processes in BrCa. Proteasome inhibitors (PIs) emerged as promising anticancer agents for several human malignancies, including BrCa, inhibiting the function of the proteasome. Aiming to shed light on the miRNA regulatory effect in BrCa after treatment with PIs, we used two PIs, namely bortezomib and carfilzomib. MATERIALS AND METHODS Four BrCa cell lines of distinct molecular subtypes were treated with these PIs. Cell viability and IC50 concentrations were determined. Total RNA was extracted, polyadenylated, and reversely transcribed. Next, the levels of specific miRNAs with a significant role in BrCa were determined using relative quantification, and their regulatory effect was assessed. RESULTS High heterogeneity was discovered in the levels of miRNAs in the four cell lines, after treatment. The miRNA levels fluctuate with distinct patterns, in 24, 48, or 72 hours. Interestingly, miR-1-3p, miR-421-3p, and miR-765-3p appear as key molecules, as they were found deregulated, in almost all combinations of cell lines and PIs. In the SK-BR-3 cell line, the majority of the miRNAs were significantly downregulated in treated compared to untreated cells, with miR-21-5p being the only one upregulated. Finally, various significant biological processes, molecular functions, and pathways were predicted to be affected. CONCLUSIONS The diversity of pathways predicted to be affected by the diversity in miRNA expression after treatment with PIs paves the way for the recognition of new regulatory axes in BrCa.
Collapse
Affiliation(s)
- Katerina Katsaraki
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15701, Athens, Greece
| | - Christos K Kontos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15701, Athens, Greece.
| | - Gerasimos Ardavanis-Loukeris
- First Department of Medical Oncology, "Saint Savvas" General Anticancer Hospital of Athens, 11522, Athens, Greece
| | - Alexandros A Tzovaras
- First Department of Medical Oncology, "Saint Savvas" General Anticancer Hospital of Athens, 11522, Athens, Greece
| | - Diamantis C Sideris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15701, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15701, Athens, Greece
| |
Collapse
|
16
|
Lv JT, Jiao YT, Han XL, Cao YJ, Lv XK, Du J, Hou J. Integrating p53-associated genes and infiltrating immune cell characterization as a prognostic biomarker in multiple myeloma. Heliyon 2024; 10:e30123. [PMID: 38699735 PMCID: PMC11063508 DOI: 10.1016/j.heliyon.2024.e30123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Background Tumor genetic anomalies and immune dysregulation are pivotal in the progression of multiple myeloma (MM). Accurate patient stratification is essential for effective MM management, yet current models fail to comprehensively incorporate both molecular and immune profiles. Methods We examined 776 samples from the MMRF CoMMpass database, employing univariate regression with LASSO and CIBERSORT algorithms to identify 15 p53-related genes and six immune cells with prognostic significance in MM. A p53-TIC (tumor-infiltrating immune cells) classifier was constructed by calculating scores using the bootstrap-multicox method, which was further validated externally (GSE136337) and through ten-fold internal cross-validation for its predictive reliability and robustness. Results The p53-TIC classifier demonstrated excellent performance in predicting the prognosis in MM. Specifically, patients in the p53low/TIChigh subgroup had the most favorable prognosis and the lowest tumor mutational burden (TMB). Conversely, those in the p53high/TIClow subgroup, with the least favorable prognosis and the highest TMB, were predicted to have the best anti-PD1 and anti-CTLA4 response rate (40 %), which can be explained by their higher expression of PD1 and CTLA4. The three-year area under the curve (AUC) was 0.80 in the total sample. Conclusions Our study highlights the potential of an integrated analysis of p53-associated genes and TIC in predicting prognosis and aiding clinical decision-making in MM patients. This finding underscores the significance of comprehending the intricate interplay between genetic abnormalities and immune dysfunction in MM. Further research into this area may lead to the development of more effective treatment strategies.
Collapse
Affiliation(s)
- Jun-Ting Lv
- Zhuhai Hospital of Integrated Traditional Chinese & Western Medicine, 519000, China
| | - Yu-Tian Jiao
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin-Le Han
- Department of Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yang-Jia Cao
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China
| | - Xu-Kun Lv
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Department of Hematology, Punan Hospital, Pudong New District, Shanghai, 200011, China
| | - Jian Hou
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
17
|
Guo Y, Wu D, Li X, Wang J, Li H, Li Y, Luo D, Yi F, Zhang D. Proximal tubular MBD2 promotes autophagy to drive the progression of AKI caused by vancomycin via regulation of miR-597-5p/S1PR1 axis. FASEB J 2024; 38:e23562. [PMID: 38578557 DOI: 10.1096/fj.202301500r] [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: 07/23/2023] [Revised: 02/03/2024] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Our recent investigation has indicated that the global deletion of MBD2 can mitigate the progression of AKI induced by VAN. Nevertheless, the role and regulatory mechanisms of proximal tubular MBD2 in this pathophysiological process have yet to be elucidated. Our preceding investigation revealed that autophagy played a crucial role in advancing AKI induced by VAN. Consequently, we postulated that MBD2 present in the proximal tubule could upregulate the autophagic process to expedite the onset of AKI. In the present study, we found for the first time that MBD2 mediated the autophagy production induced by VAN. Through the utilization of miRNA chip analysis, we have mechanistically demonstrated that MBD2 initiates the activation of miR-597-5p through promoter demethylation. This process leads to the suppression of S1PR1, which results in the induction of autophagy and apoptosis in renal tubular cells. Besides, PT-MBD2-KO reduced autophagy to attenuate VAN-induced AKI via regulation of the miR-597-5p/S1PR1 axis, which was reversed by rapamycin. Finally, the overexpression of MBD2 aggravated the diminished VAN-induced AKI in autophagy-deficient mice (PT-Atg7-KO). These data demonstrate that proximal tubular MBD2 facilitated the process of autophagy via the miR-597-5p/S1PR1 axis and subsequently instigated VAN-induced AKI through the induction of apoptosis. The potentiality of MBD2 being a target for AKI was established.
Collapse
Affiliation(s)
- Yong Guo
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Organ Procurement Organization, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dengke Wu
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaozhou Li
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Wang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huiling Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yijian Li
- Department of Urinary Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan Luo
- Department of Emergency Medicine, Yueyang Central Hospital, Yueyang, Hunan, China
| | - Feng Yi
- Department of Emergency Medicine, Yueyang Central Hospital, Yueyang, Hunan, China
| | - Dongshan Zhang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
18
|
Mao J, Chen R, Xue L, Zhu Y, Zhao L, Wang J. Metformin and chidamide synergistically suppress multiple myeloma progression and enhance lenalidomide/bortezomib sensitivity. ENVIRONMENTAL TOXICOLOGY 2024; 39:2452-2465. [PMID: 38251764 DOI: 10.1002/tox.24093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 12/01/2023] [Indexed: 01/23/2024]
Abstract
Multiple myeloma (MM) is a common hematological malignancy, and patients with MM are recommended to take immunomodulatory drugs such as lenalidomide along with proteasome inhibitors such as bortezomib to extend survival. However, drug resistance influences the efficacy of treatment for MM. In our study, we found that metformin and chidamide both suppressed MM cell growth in a concentration- and time-dependent way (p < .001). Moreover, combined therapy with metformin and chidamide exhibited enhanced inhibition of the growth of MM cells compared with monotherapy (p < .05). Additionally, the triple-drug combination of metformin and chidamide with lenalidomide or bortezomib was used to stimulate the MM cells, and the results revealed that metformin and chidamide treatment sensitized MM cells to lenalidomide and bortezomib. As a result, the apoptosis (p < .001) together with cell cycle arrest at G0/G1 phase (p < .05) was stimulated by lenalidomide and bortezomib, and showed significant elevation in the triple-drug combination group compared with the lenalidomide or bortezomib treatment alone group (p < .05). Furthermore, the impacts of different drugs on glycolysis in MM cells were examined. We found that metformin and chidamide combined treatment significantly promoted glucose uptake and reduced energy production in MM cells treated with lenalidomide and bortezomib (p < .001), suggesting that metformin and chidamide affected glycolysis in MM cells and enhanced the sensitivity of lenalidomide and bortezomib in MM by regulating glucose metabolism. In conclusion, metformin and chidamide synergistically hindered MM cell growth and sensitized cells to lenalidomide/bortezomib. The findings of this study might provide novel clues to improve MM therapy.
Collapse
Affiliation(s)
- Jianping Mao
- Department of Hematology, the First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Ran Chen
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lianguo Xue
- Department of Hematology, the First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Yuanxin Zhu
- Department of Hematology, the First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Lidong Zhao
- Department of Hematology, the First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Juan Wang
- Department of Pediatrics, the First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| |
Collapse
|
19
|
Zeng RY, Jin HY, Peng YB, Wang WJ, Cao YP, Peng HZ, Qiu ZC, Lai SQ, Wan L. miR-200a-3p inhibits the PDGF-BB-induced proliferation of VSMCs by affecting their phenotype-associated proteins. J Biochem Mol Toxicol 2024; 38:e23675. [PMID: 38488158 DOI: 10.1002/jbt.23675] [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: 08/22/2022] [Revised: 09/19/2023] [Accepted: 02/23/2024] [Indexed: 03/19/2024]
Abstract
Accumulating evidence shows that the abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) can significantly affect the long-term prognosis of coronary artery bypass grafting. This study aimed to explore the factors affecting the proliferation, migration, and phenotypic transformation of VSMCs. First, we stimulated VSMCs with different platelet-derived growth factor-BB (PDGF-BB) concentrations, analyzed the expression of phenotype-associated proteins by Western blotting, and examined cell proliferation by scratch wound healing and the 5-ethynyl-2-deoxyuridine (EdU) assay. VSMC proliferation was induced most by PDGF-BB treatment at 20 ng/mL. miR-200a-3p decreased significantly in A7r5 cells stimulated with PDGF-BB. The overexpression of miR-200a-3p reversed the downregulation of α-SMA (p < 0.001) and the upregulation of vimentin (p < 0.001) caused by PDGF-BB. CCK8 and EdU analyses showed that miR-200a-3p overexpression could inhibit PDGF-BB-induced cell proliferation (p < 0.001). However, flow cytometric analysis showed that it did not significantly increase cell apoptosis. Collectively, the overexpression of miR-200a-3p inhibited the proliferation and migration of VSMCs induced by PDGF-BB, partly by affecting phenotypic transformation-related proteins, providing a new strategy for relieving the restenosis of vein grafts.
Collapse
Affiliation(s)
- Rui-Yuan Zeng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hong-Yi Jin
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yong-Bo Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wen-Jun Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuan-Ping Cao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Han-Zhi Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhi-Cong Qiu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Song-Qing Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Li Wan
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Institute of Cardiovascular Surgical Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
20
|
Zuo Z, Li Q, Zhou S, Yu R, Wu C, Chen J, Xiao Y, Chen H, Song J, Pan Y, Wang W. Berberine ameliorates contrast-induced acute kidney injury by regulating HDAC4-FoxO3a axis-induced autophagy: In vivo and in vitro. Phytother Res 2024; 38:1761-1780. [PMID: 37922559 DOI: 10.1002/ptr.8059] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/30/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
In hospitals, contrast-induced acute kidney injury (CI-AKI) is a major cause of renal failure. This study evaluates berberine's (BBR) renal protection and its potential HDAC4 mechanism. CI-AKI in rats was induced with 10 mL kg-1 ioversol. Rats were divided into five groups: Ctrl, BBR, CI-AKI, CI-AKI + BBR, and CI-AKI + Tasq. The renal function of CI-AKI rats was determined by measuring serum creatinine and blood urea nitrogen. Histopathological changes and apoptosis of renal tubular epithelial cells were observed by HE and terminal deoxynucleotidyl transferase (TdTase)-mediated dUTP-biotin nick end labeling (TUNEL) staining. Transmission electron microscopy was used to observe autophagic structures. In vitro, a CI-AKI cell model was created with ioversol-treated HK-2 cells. Treatments included BBR, Rapa, HCQ, and Tasq. Analyses focused on proteins and genes associated with kidney injury, apoptosis, autophagy, and the HDAC4-FoxO3a axis. BBR showed significant protective effects against CI-AKI both in vivo and in vitro. It inhibited apoptosis by increasing Bcl-2 protein levels and decreasing Bax levels. BBR also activated autophagy, as indicated by changes in autophagy-related proteins and autophagic flux. The study further revealed that the contrast agent ioversol increased the expression of HDAC4, which led to elevated levels of phosphorylated FoxO3a (p-FoxO3a) and acetylated FoxO3a (Ac-FoxO3a). However, BBR inhibited HDAC4 expression, resulting in decreased levels of p-FoxO3a and Ac-FoxO3a. This activation of autophagy-related genes, regulated by the transcription factor FoxO3a, played a role in BBR's protective effects. BBR, a traditional Chinese medicine, shows promise against CI-AKI. It may counteract CI-AKI by modulating HDAC4 and FoxO3a, enhancing autophagy, and limiting apoptosis.
Collapse
Affiliation(s)
- Zhi Zuo
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University/Jiangsu Province Hospital, Nanjing, China
| | - Qingju Li
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| | - Suqin Zhou
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Ran Yu
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| | - Caixia Wu
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Jiajia Chen
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Yao Xiao
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- Jiangsu College of Nursing, Huai'an, China
| | - Haoyu Chen
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Jian Song
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Yan Pan
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
| | - Wanpeng Wang
- Lianshui People's Hospital, Affiliated Kangda College of Nanjing Medical University, Huai'an, China
- School of Clinical Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Jiangsu College of Nursing, Huai'an, China
| |
Collapse
|
21
|
Kozalak G, Koşar A. Autophagy-related mechanisms for treatment of multiple myeloma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:838-857. [PMID: 38239705 PMCID: PMC10792488 DOI: 10.20517/cdr.2023.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Multiple myeloma (MM) is a type of hematological cancer that occurs when B cells become malignant. Various drugs such as proteasome inhibitors, immunomodulators, and compounds that cause DNA damage can be used in the treatment of MM. Autophagy, a type 2 cell death mechanism, plays a crucial role in determining the fate of B cells, either promoting their survival or inducing cell death. Therefore, autophagy can either facilitate the progression or hinder the treatment of MM disease. In this review, autophagy mechanisms that may be effective in MM cells were covered and evaluated within the contexts of unfolded protein response (UPR), bone marrow microenvironment (BMME), drug resistance, hypoxia, DNA repair and transcriptional regulation, and apoptosis. The genes that are effective in each mechanism and research efforts on this subject were discussed in detail. Signaling pathways targeted by new drugs to benefit from autophagy in MM disease were covered. The efficacy of drugs that regulate autophagy in MM was examined, and clinical trials on this subject were included. Consequently, among the autophagy mechanisms that are effective in MM, the most suitable ones to be used in the treatment were expressed. The importance of 3D models and microfluidic systems for the discovery of new drugs for autophagy and personalized treatment was emphasized. Ultimately, this review aims to provide a comprehensive overview of MM disease, encompassing autophagy mechanisms, drugs, clinical studies, and further studies.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
22
|
Kulig P, Łuczkowska K, Bakinowska E, Baumert B, Machaliński B. Epigenetic Alterations as Vital Aspects of Bortezomib Molecular Action. Cancers (Basel) 2023; 16:84. [PMID: 38201512 PMCID: PMC10778101 DOI: 10.3390/cancers16010084] [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: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Bortezomib (BTZ) is widely implemented in the treatment of multiple myeloma (MM). Its main mechanism of action is very well established. BTZ selectively and reversibly inhibits the 26S proteasome. More precisely, it interacts with the chymotryptic site of the 20S proteasome and therefore inhibits the degradation of proteins. This results in the intracellular accumulation of misfolded or otherwise defective proteins leading to growth inhibition and apoptosis. As well as interfering with the ubiquitin-proteasome complex, BTZ elicits various epigenetic alterations which contribute to its cytotoxic effects as well as to the development of BTZ resistance. In this review, we summarized the epigenetic alterations elicited by BTZ. We focused on modifications contributing to the mechanism of action, those mediating drug-resistance development, and epigenetic changes promoting the occurrence of peripheral neuropathy. In addition, there are therapeutic strategies which are specifically designed to target epigenetic changes. Herein, we also reviewed epigenetic agents which might enhance BTZ-related cytotoxicity or restore the sensitivity to BTZ of resistant clones. Finally, we highlighted putative future perspectives regarding the role of targeting epigenetic changes in patients exposed to BTZ.
Collapse
Affiliation(s)
- Piotr Kulig
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.K.); (K.Ł.); (E.B.)
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.K.); (K.Ł.); (E.B.)
| | - Estera Bakinowska
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.K.); (K.Ł.); (E.B.)
| | - Bartłomiej Baumert
- Department of Hematology and Transplantology, Pomeranian Medical University, 71-252 Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, 70-111 Szczecin, Poland; (P.K.); (K.Ł.); (E.B.)
- Department of Hematology and Transplantology, Pomeranian Medical University, 71-252 Szczecin, Poland
| |
Collapse
|
23
|
Liu X, Liu Z, Liu Y, Wang N. ATG9A modulated by miR-195-5p can boost the malignant progression of cervical cancer cells. Epigenetics 2023; 18:2257538. [PMID: 37782756 PMCID: PMC10547073 DOI: 10.1080/15592294.2023.2257538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/01/2023] [Indexed: 10/04/2023] Open
Abstract
Cervical cancer (CC) is a major public health problem, and its molecular mechanism requires further investigation. The goal of this study was to determine the role of miR-195-5p and the autophagy-related protein ATG9A in tumour metastasis, epithelial - mesenchymal transition (EMT), apoptosis, and autophagy of CC cells. Using bioinformatics analysis, we predicted ATG9A as a downstream target gene of miR-195-5p, an integral membrane protein required for autophagosome formation and involved in tumorigenesis. Next, western blotting and Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) showed that upregulation of miR-195-5p decreased protein and mRNA expression of ATG9A, and downregulation of miR-195-5p promoted ATG9A protein and mRNA expression. In addition, detection of the dual luciferase reporter gene further indicated ATG9A is a direct downstream target gene of miR-195-5p. Finally, the effects of miR-195-5p and ATG9A on CC cell proliferation, migration, invasion, EMT, autophagy, and apoptosis were evaluated in vitro. Our results showed that upregulation of miR-195-5p not only inhibits proliferation, migration, and the EMT of CC cells, but also induces apoptosis and autophagy. Conversely, downregulation of miR-195-5p increased malignant metastasis and the EMT of CC cells, and inhibited apoptosis as well as autophagy. In addition, miR-195-5p targeted and negatively regulated ATG9A, and rescue experiments suggested that overexpression of ATG9A could partially abolish miR-195-5p-mediated suppression of CC cells. Our findings improve our understanding of the mechanism of action of miR-195-5p in the malignant behaviour of CC. miR-195-5p is likely to be a promising cancer suppressor gene, which provides clinical evidence for targeted therapy of CC.
Collapse
Affiliation(s)
- Xiaomin Liu
- Department of Gynecology, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, China
| | - Zhen Liu
- Department of Gynecology, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, China
| | - Yonggang Liu
- Department of Gynecology, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, China
| | - Ning Wang
- Department of Gynecology, The Second Hospital of Dalian Medical University, DaLian, China
| |
Collapse
|
24
|
Long ZJ, Wang JD, Qiu SX, Zhang Y, Wu SJ, Lei XX, Huang ZW, Chen JJ, Yang YL, Zhang XZ, Liu Q. Dietary γ-mangostin triggers immunogenic cell death and activates cGAS signaling in acute myeloid leukemia. Pharmacol Res 2023; 197:106973. [PMID: 37898441 DOI: 10.1016/j.phrs.2023.106973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Immunogenic cell death (ICD), one of cell-death types through release of damage-associated molecular patterns from dying tumor cells, activates tumor-specific immune response and elicits anti-tumor immunity by traditional radiotherapy and chemotherapy. However, whether natural products could induce ICD in leukemia is not elucidated. Here, we report dietary γ-mangostin eradicates murine primary leukemic cells and prolongs the survival of leukemic mice. As well, it restrains primary leukemic cells and CD34+ leukemic progenitor cells from leukemia patients. Strikingly, γ-mangostin attenuates leukemic cells by inducing ICD as characterized by expression of HSP90B1, ANXA1 and IL1B. Additionally, γ-mangostin accelerates cytoplasmic chromatin fragments generation, promoting DNA damage response, and enhances cGAS activation, leading to up-regulation of chemokines. Meanwhile, it induces HDAC4 degradation and acetylated histone H3 accumulation, which promotes chemokines transcription. Ultimately, CD8+ T cell is activated and recruited by γ-mangostin-induced chemokines in the microenvironment. Our study identifies γ-mangostin triggers ICD and activates cGAS signaling through DNA damage response and epigenetic modification. Therefore, dietary γ-mangostin would act as a potential agent to provoke anti-tumor immunity in the prevention and treatment of leukemia.
Collapse
Affiliation(s)
- Zi-Jie Long
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, China; Institute of Hematology, Sun Yat-sen University, China; Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, China.
| | - Jun-Dan Wang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, China; Institute of Hematology, Sun Yat-sen University, China
| | - Sheng-Xiang Qiu
- State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, China
| | - Yi Zhang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, China; Institute of Hematology, Sun Yat-sen University, China
| | - Si-Jin Wu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, China
| | - Xin-Xing Lei
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, China
| | - Ze-Wei Huang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, China; Institute of Hematology, Sun Yat-sen University, China
| | - Jia-Jie Chen
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, China; Institute of Hematology, Sun Yat-sen University, China
| | - Yong-Liang Yang
- Center for Molecular Medicine, School of Life Science and Biotechnology, Dalian University of Technology, China
| | - Xiang-Zhong Zhang
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, China; Institute of Hematology, Sun Yat-sen University, China.
| | - Quentin Liu
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, China; Institute of Hematology, Sun Yat-sen University, China; Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, China.
| |
Collapse
|
25
|
Ahmad MS, Braoudaki M, Patel H, Ahmad I, Shagufta, Siddiqui SS. Novel Siglec-15-Sia axis inhibitor leads to colorectal cancer cell death by targeting miR-6715b-3p and oncogenes. Front Immunol 2023; 14:1254911. [PMID: 37869015 PMCID: PMC10587484 DOI: 10.3389/fimmu.2023.1254911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Siglecs are well known immunotherapeutic targets in cancer. Current checkpoint inhibitors have exhibited limited efficacy, prompting a need for novel therapeutics for targets such as Siglec-15. Presently, small molecule inhibitors targeting Siglec-15 are not explored alongside characterised regulatory mechanisms involving microRNAs in CRC progression. Therefore, a small molecule inhibitor to target Siglec-15 was elucidated in vitro and microRNA mediated inhibitor effects were investigated. Our research findings demonstrated that the SHG-8 molecule exerted significant cytotoxicity on cell viability, migration, and colony formation, with an IC50 value of approximately 20µM. SHG-8 exposure induced late apoptosis in vitro in SW480 CRC cells. Notably, miR-6715b-3p was the most upregulated miRNA in high-throughput sequencing, which was also validated via RT-qPCR. MiR-6715b-3p may regulate PTTG1IP, a potential oncogene which was validated via RT-qPCR and in silico analysis. Additionally, molecular docking studies revealed SHG-8 interactions with the Siglec-15 binding pocket with the binding affinity of -5.4 kcal/mol, highlighting its role as a small molecule inhibitor. Importantly, Siglec-15 and PD-L1 are expressed on mutually exclusive cancer cell populations, suggesting the potential for combination therapies with PD-L1 antagonists.
Collapse
Affiliation(s)
- Mohammed Saqif Ahmad
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Maria Braoudaki
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Hershna Patel
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Irshad Ahmad
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates
| | - Shagufta
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates
| | - Shoib Sarwar Siddiqui
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
26
|
Xu Y, He Z, Du J, Chen Z, Creemers JWM, Wang B, Li F, Wang Y. Epigenetic modulations of immune cells: from normal development to tumor progression. Int J Biol Sci 2023; 19:5120-5144. [PMID: 37928272 PMCID: PMC10620821 DOI: 10.7150/ijbs.88327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/21/2023] [Indexed: 11/07/2023] Open
Abstract
The dysfunction of immune cell development often impairs immunological homeostasis, thus causing various human diseases. Accumulating evidence shows that the development of different immune cells from hematopoietic stem cells are highly fine-tuned by different epigenetic mechanisms including DNA methylation, histone modifications, chromatin remodeling and RNA-related regulations. Understanding how epigenetic regulators modulate normal development of immune cells contributes to the identification of new strategies for various diseases. Here, we review recent advances suggesting that epigenetic modulations can orchestrate immune cell development and functions through their impact on critical gene expression. We also discuss the aberrations of epigenetic modulations in immune cells that influence tumor progression, and the fact that underlying mechanisms affect how epigenetic drugs interfere with tumor progression in the clinic.
Collapse
Affiliation(s)
- Yuanchun Xu
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
- Department of nursing, Daping Hospital, Army Medical University, Chongqing, China
| | - Zongsheng He
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Du
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ziqiang Chen
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | | | - Bin Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Fan Li
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yaling Wang
- Department of nursing, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
27
|
Hao S, Yao Z, Liu Y. Hsa_circ_0000106 Acts as a Tumor Promoter in Pancreatic Cancer by Targeting the MiR-455-3p/HDAC4. Horm Metab Res 2023; 55:722-732. [PMID: 37553012 DOI: 10.1055/a-2125-7018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Circular RNAs (circRNAs) frequently participate in pancreatic cancer (PC) progression. This study focuses on circ_0000106, a novel circRNA, and its potential function in PC development. Circ_00001106, miR-455-3p, and HDAC4 expression levels in PC were determined using qRT-PCR and immunoblotting. RNA immunoprecipitation and dual-luciferase reporter assays were performed to verify their binding interactions. Loss-of-function assays, including CCK-8, colony formation, and transwell assays, were used to estimate the proliferative and migratory properties of PC cells. A nude mouse model was constructed to assess the influence of circ_0000106 on tumor formation in vivo. A pronounced elevation of circ_0000106 and HDAC4 and a reduction of miR-455-3p in PC were observed. Circ_0000106 was prone to binding to miR-455-3p, and miR-455-3p further targeted HDAC4. Functionally, the proliferative and migratory properties of PC cells were dampened by the loss of circ_0000106 or HDAC4 and could be potentiated by miR-455-3p inhibition. Moreover, the knockdown of circ_0000106 delayed tumor growth in vivo. Additionally, the downregulation of miR-455-3p attenuated the repressive effects of circ_0000106 deficiency on PC cell migration and proliferation. Loss of HDAC4 exerted similar mitigative effects on miR-455-3p downregulation-stimulated PC cells. In conclusion, circ_0000106 promotes tumor migration and growth in PC by targeting the miR-455-3p/HDAC4 axis. These results suggest that the circ_0000106/miR-455-3p/HDAC4 network could be regarded as a latent target for PC treatment.
Collapse
Affiliation(s)
- Shunxin Hao
- Department of General Surgery, Wuhan University of Science and Technology Hospital, Wuhan, China
| | - Zhi Yao
- Department of General Surgery, Wuhan University of Science and Technology Hospital, Wuhan, China
| | - Yifeng Liu
- Department of General Surgery, Wuhan University of Science and Technology Hospital, Wuhan, China
| |
Collapse
|
28
|
Zhu G, Jin L, Shen W, Zhao M, Liu N. Intratumor microbiota: Occult participants in the microenvironment of multiple myeloma. Biochim Biophys Acta Rev Cancer 2023; 1878:188959. [PMID: 37488050 DOI: 10.1016/j.bbcan.2023.188959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
More recently, microbiota was detected in several tumorous tissues including multiple myeloma (MM), but the roles of which is still under-studied as paucity of research on tumor biology. Moreover, we also detected the presence of microbiota in the bone marrow of patients with MM by 2bRAD-M sequencing technology, which is an incurable hematological malignancy characterized by accumulation of abnormal plasma cells in the bone marrow. However, the roles of intratumor microbiota in tumor disease remains poorly understood. In this review, we critically reviewed recent literature about microbiota in the tumorigenesis and progression of MM. Importantly, we proposed that the emergence of microbiota in the microenvironment of multiple myeloma may be attributed to microbial dysbiosis and impaired intestinal barrier, due to the increased prevalence of MM in patients with obesity and diabetes, of which the characteristic phenotype is gut microbial dysbiosis and impaired intestinal barrier. When the intestinal barrier is damaged, dysbiotic microbiota and their metabolites, as well as dysregulated immune cells, may participate in the reshaping of the local immune microenvironment, and play pivotal roles in the tumorigenesis and development of multiple myeloma, probably by migrating to the bone marrow microenvironment from intestine. We also discuss the emerging microbiological manipulation strategies to improve long-term outcomes of MM, as well as the prospective of the state-of-the-art techniques to advance our knowledge about the biological implication in the microbiome in MM.
Collapse
Affiliation(s)
- Gengjun Zhu
- Central Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Lifang Jin
- Department of Oncology and Hematology, The Second Hospital of Jilin University, Changchun, China
| | - Weizhang Shen
- Department of Oncology and Hematology, The Second Hospital of Jilin University, Changchun, China
| | - Meng Zhao
- Department of Oncology and Hematology, The Second Hospital of Jilin University, Changchun, China
| | - Ning Liu
- Central Laboratory, The Second Hospital of Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China.
| |
Collapse
|
29
|
Elkady MA, Yehia AM, Elsakka EGE, Abulsoud AI, Abdelmaksoud NM, Elshafei A, Elkhawaga SY, Ismail A, Mokhtar MM, El-Mahdy HA, Hegazy M, Elballal MS, Mohammed OA, El-Husseiny HM, Midan HM, El-Dakroury WA, Zewail MB, Abdel Mageed SS, Doghish AS. miRNAs driving diagnosis, progression, and drug resistance in multiple myeloma. Pathol Res Pract 2023; 248:154704. [PMID: 37499518 DOI: 10.1016/j.prp.2023.154704] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
Multiple myeloma (MM) is a tumor of transformed plasma cells. It's the second most common hematologic cancer after non-Hodgkin lymphoma. MM is a complex disease with many different risk factors, including ethnicity, race, and epigenetics. The microRNAs (miRNAs) are a critical epigenetic factor in multiple myeloma, influencing key aspects such as pathogenesis, prognosis, and resistance to treatment. They have the potential to assist in disease diagnosis and modulate the resistance behavior of MM towards therapeutic regimens. These characteristics could be attributed to the modulatory effects of miRNAs on some vital pathways such as NF-KB, PI3k/AKT, and P53. This review discusses the role of miRNAs in MM with a focus on their role in disease progression, diagnosis, and therapeutic resistance.
Collapse
Affiliation(s)
- Mohamed A Elkady
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Amr Mohamed Yehia
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Ahmed Elshafei
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Mahmoud Mohamed Mokhtar
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Maghawry Hegazy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Osama A Mohammed
- Department of Clinical Pharmacology, Faculty of Medicine, Bisha University, Bisha 61922, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Hussein M El-Husseiny
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt; Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Heba M Midan
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Moataz B Zewail
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
30
|
Hu C, Liu T, Zhang W, Sun Y, Jiang D, Zhang X, Liu Y, Mao S, Xu Y, Pan J, Wang J, Huang Y, Yang S, Yang K. miR-145 inhibits aerobic glycolysis and cell proliferation of cervical cancer by acting on MYC. FASEB J 2023; 37:e22839. [PMID: 36946075 DOI: 10.1096/fj.202201189rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/04/2023] [Accepted: 02/13/2023] [Indexed: 03/23/2023]
Abstract
Nearly half a million women are diagnosed with cervical cancer (CC) each year, with the incidence of CC stabilizing or rising in low-income and middle-income countries. Cancer cells use metabolic reprogramming to meet the needs of rapid proliferation, known as the Warburg effect, but the mechanism of the Warburg effect in CC remains unclear. microRNAs (miRNAs) have a wide range of effects on gene expression and diverse modes of action, and they regulate genes for metabolic reprogramming. Dysregulation of miRNA expression leads to metabolic abnormalities in tumor cells and promotes tumorigenesis and tumor progression. In this study, we found that miR-145 was negatively correlated with metabolic reprogramming-related genes and prevented the proliferation and metastasis of CC cell lines by impeding aerobic glycolysis. A dual-luciferase reporter assay showed that miR-145 can bind to the 3'-untranslated region (3'-UTR) of MYC. Chromatin Immunoprecipitation-quantitative real-time PCR indicated that MYC was involved in the regulation of glycolysis-related genes. In addition, miR-145 mimics significantly suppressed the growth of CC cell xenograft tumor, prolonged the survival time of mice, and dramatically silenced the expression of tumor proliferation marker Ki-67. Therefore, the results suggested that miR-145 affects aerobic glycolysis through MYC, which may be a potential target for the treatment of CC.
Collapse
Affiliation(s)
- Chenchen Hu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Tianyue Liu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Wenxin Zhang
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Yuanjie Sun
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Dongbo Jiang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Xiyang Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Yang Liu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Siyi Mao
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Yiming Xu
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Jingyu Pan
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Jing Wang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Yinan Huang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Shuya Yang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Kun Yang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
31
|
Peng B, Li J, Yan Y, Liu Y, Liang Q, Liu W, Thakur A, Zhang K, Xu Z, Wang J, Zhang F. Non-coding RNAs: The recently accentuated molecules in the regulation of cell autophagy for ovarian cancer pathogenesis and therapeutic response. Front Pharmacol 2023; 14:1162045. [PMID: 37063265 PMCID: PMC10102359 DOI: 10.3389/fphar.2023.1162045] [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: 02/09/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Autophagy is a self-recycling and conserved process, in which the senescent cytoplasmic components are degraded in cells and then recycled to maintain homeostatic balance. Emerging evidence has suggested the involvement of autophagy in oncogenesis and progression of various cancers, such as ovarian cancer (OC). Meanwhile, the non-coding RNAs (ncRNAs) frequently regulate the mRNA transcription and other functional signaling pathways in cell autophagy, displaying promising roles in human cancer pathogenesis and therapeutic response. This article mainly reviews the cutting-edge research advances about the interactions between ncRNAs and autophagy in OC. This review not only summarizes the underlying mechanisms of dynamic ncRNA-autophagy association in OC, but also discusses their prognostic implications and therapeutic biomarkers. The aim of this review was to provide a more in-depth knowledge framework exploring the ncRNA-autophagy crosstalk and highlight the promising treatment strategies for OC patients.
Collapse
Affiliation(s)
- Bi Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Li
- Department of Pharmacy, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Orthopedic Surgery, The Second Hospital University of South China, Hengyang, Hunan, China
| | - Abhimanyu Thakur
- Ben May Department for Cancer Research, Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, Medical Research Institute, Southwest University, Chongqing, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Wang
- Department of Pharmacy, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Fan Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
32
|
Ma L, Gong Q, Chen Y, Luo P, Chen J, Shi C. Targeting positive cofactor 4 induces autophagic cell death in MYC-expressing diffuse large B-cell lymphoma. Exp Hematol 2023; 119-120:42-57.e4. [PMID: 36642374 DOI: 10.1016/j.exphem.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
MYC-expressing diffuse large B-cell lymphoma (DLBCL) is one of the refractory lymphomas. Currently, the pathogenesis of MYC-expressing DLBCL is still unclear, and there is a lack of effective therapy. We characterized positive cofactor 4 (PC4) as an upstream regulator of c-Myc, and PC4 is overexpressed in DLBCL and is closely related to clinical staging, prognosis, and c-Myc expression. Furthermore, our in vivo and in vitro studies revealed that PC4 knockdown can induce autophagic cell death and enhance the therapeutic effect of doxorubicin in MYC-expressing DLBCL. Inhibition of c-Myc-mediated aerobic glycolysis and activation of the AMPK/mTOR signaling pathway are responsible for the autophagic cell death induced by PC4 knockdown in MYC-expressing DLBCL. Using dual-luciferase reporter assay and electrophoretic mobility shift assay assays, we also found that PC4 exerts its oncogenic functions by directly binding to c-Myc promoters. To sum up, our study provides novel insights into the functions and mechanisms of PC4 in MYC-expressing DLBCL and suggests that PC4 may be a promising therapeutic target for MYC-expressing DLBCL.
Collapse
Affiliation(s)
- Le Ma
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Hematology, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing 400038, China
| | - Qiang Gong
- Department of Hematology, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing 400038, China
| | - Yan Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peng Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing 400038, China.
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
33
|
Cuttini E, Goi C, Pellarin E, Vida R, Brancolini C. HDAC4 in cancer: A multitasking platform to drive not only epigenetic modifications. Front Mol Biosci 2023; 10:1116660. [PMID: 36762207 PMCID: PMC9902726 DOI: 10.3389/fmolb.2023.1116660] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Controlling access to genomic information and maintaining its stability are key aspects of cell life. Histone acetylation is a reversible epigenetic modification that allows access to DNA and the assembly of protein complexes that regulate mainly transcription but also other activities. Enzymes known as histone deacetylases (HDACs) are involved in the removal of the acetyl-group or in some cases of small hydrophobic moieties from histones but also from the non-histone substrate. The main achievement of HDACs on histones is to repress transcription and promote the formation of more compact chromatin. There are 18 different HDACs encoded in the human genome. Here we will discuss HDAC4, a member of the class IIa family, and its possible contribution to cancer development.
Collapse
Affiliation(s)
- Emma Cuttini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Camilla Goi
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Ester Pellarin
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Riccardo Vida
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Claudio Brancolini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy,Laboratory of Epigenomics, Department of Medicine, Università degli Studi di Udine, Udine, Italy,*Correspondence: Claudio Brancolini,
| |
Collapse
|
34
|
Yin Y, Zhou Y, Yang X, Xu Z, Yang B, Luo P, Yan H, He Q. The participation of non-canonical autophagic proteins in the autophagy process and their potential as therapeutic targets. Expert Opin Ther Targets 2023; 27:71-86. [PMID: 36735300 DOI: 10.1080/14728222.2023.2177151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Autophagy is a conserved catabolic process that helps recycle intracellular components to maintain homeostasis. The completion of autophagy requires the synergistic effect of multiple canonical autophagic proteins. Defects in autophagy machinery have been reported to promote diseases, rendering autophagy a bone fide health-modifying agent. However, the clinical implication of canonical pan-autophagic activators or inhibitors has often led to undesirable side effects, making it urgent to find a safer autophagy-related therapeutic target. The discovery of non-canonical autophagic proteins has been found to specifically affect the development of diseases without causing a universal impact on autophagy and has shed light on finding a safer way to utilize autophagy in the therapeutic context. AREAS COVERED This review summarizes recently discovered non-canonical autophagic proteins, how these proteins influence autophagy, and their potential therapeutic role in the disease due to their interaction with autophagy. EXPERT OPINION Several therapies have been studied thus far and continued research is needed to identify the potential that non-canonical autophagic proteins have for treating certain diseases. In the meantime, continue to uncover new non-canonical autophagic proteins and examine which are likely to have therapeutic implications.
Collapse
Affiliation(s)
- Yiming Yin
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yourong Zhou
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Alotaibi F. Exosomal microRNAs in cancer: Potential biomarkers and immunotherapeutic targets for immune checkpoint molecules. Front Genet 2023; 14:1052731. [PMID: 36873941 PMCID: PMC9982116 DOI: 10.3389/fgene.2023.1052731] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/07/2023] [Indexed: 02/19/2023] Open
Abstract
Exosomes are small extracellular vesicles with a lipid bilayer structure secreted from different cell types which can be found in various body fluids including blood, pleural fluid, saliva and urine. They carry different biomolecules including proteins, metabolites, and amino acids such as microRNAs which are small non-coding RNAs that regulate gene expression and promote cell-to-cell communication. One main function of the exosomal miRNAs (exomiRs) is their role in cancer pathogenesis. Alternation in exomiRs expression could indicate disease progression and can regulate cancer growth and facilitate drug response/resistance. It can also influence the tumour microenvironment by controlling important signaling that regulating immune checkpoint molecules leading to activation of T cell anti-tumour immunity. Therefore, they can be used as potential novel cancer biomarkers and innovative immunotherapeutic agents. This review highlights the use of exomiRs as potential reliable biomarkers for cancer diagnosis, treatment response and metastasis. Finally, discuses their potential as immunotherapeutic agents to regulate immune checkpoint molecules and promote T cell anti-tumour immunity.
Collapse
Affiliation(s)
- Faizah Alotaibi
- College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
36
|
Lu Y, Chen X, Liu X, Shi Y, Wei Z, Feng L, Jiang Q, Ye W, Sasaki T, Fukunaga K, Ji Y, Han F, Lu YM. Endothelial TFEB signaling-mediated autophagic disturbance initiates microglial activation and cognitive dysfunction. Autophagy 2023:1-18. [PMID: 36588318 DOI: 10.1080/15548627.2022.2162244] [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] [Indexed: 01/03/2023] Open
Abstract
Cognitive impairment caused by systemic chemotherapy is a critical question that perplexes the effective implementation of clinical treatment, but related molecular events are poorly understood. Herein, we show that bortezomib exposure leads to microglia activation and cognitive impairment, this occurs along with decreased nuclear translocation of TFEB (transcription factor EB), which is linked to macroautophagy/autophagy disorder, STAT3 (signal transducer and activator of transcription 3) phosphorylation and IL23A (interleukin 23 subunit alpha) expression. Pharmacological enhancement of TFEB nuclear translocation by digoxin restores lysosomal function and reduces STAT3-dependent endothelial IL23A secretion. As a consequence, we found that brain endothelial-specific ablation of Il23a ameliorated both microglia activation and cognitive dysfunction. Thus, the endothelial TFEB-STAT3-IL23A axis in the brain represents a critical cellular event for initiating bortezomib-mediated aberrant microglial activation and synapse engulfment. Our results suggest the reversal of TFEB nuclear translocation may provide a novel therapeutic approach to prevent symptoms of cognitive dysfunction during clinical use of bortezomib.Abbreviations: AAV: adeno-associated virus; BBB: blood-brain barrier; BTZ: bortezomib; DG: digoxin; DGs: dentate gyrus; DLG4/PSD95: discs large MAGUK scaffold protein 4; HBMECs: human brain microvascular endothelial cells; HP: hippocampus; IL23A: interleukin 23 subunit alpha; MBVECs: mouse brain vascular endothelial cells; mPFC: medial prefrontal cortex; NORT: novel object recognition test; OLT: object location test; PLX5622: 6-fluoro-N-([5-fluoro-2-methoxypyridin-3-yl]methyl)-5-(5-methyl-1H-pyrrolo[2,3-b]pyridin-3- yl)methyl; PPP3/calcineurin: protein phosphatase 3; SBEs: STAT3 binding elements; shRNA: small hairpin RNA; SLC17A7/VGLUT1: solute carrier family 17 member 7; SLC32A1/VGAT: solute carrier family 32 member 1; STAT3: signal transducer and activator of transcription 3, TFEB: transcription factor EB; Ub: ubiquitin.
Collapse
Affiliation(s)
- Yaping Lu
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiang Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiuxiu Liu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Shi
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhaocong Wei
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lili Feng
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Quan Jiang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weifeng Ye
- Department of Pharmacy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yong Ji
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.,Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.,Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China.,Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying-Mei Lu
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China.,Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
37
|
Al-Odat OS, Guirguis DA, Schmalbach NK, Yao G, Budak-Alpdogan T, Jonnalagadda SC, Pandey MK. Autophagy and Apoptosis: Current Challenges of Treatment and Drug Resistance in Multiple Myeloma. Int J Mol Sci 2022; 24:ijms24010644. [PMID: 36614089 PMCID: PMC9820338 DOI: 10.3390/ijms24010644] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Over the past two decades, the natural history of multiple myeloma (MM) has evolved dramatically, owing primarily to novel agents targeting MM in the bone marrow microenvironment (BMM) pathways. However, the mechanisms of resistance acquisition remain a mystery and are poorly understood. Autophagy and apoptosis are tightly controlled processes and play a critical role in the cell growth, development, and survival of MM. Genetic instability and abnormalities are two hallmarks of MM. During MM progression, plasma malignant cells become genetically unstable and activate various signaling pathways, resulting in the overexpression of abnormal proteins that disrupt autophagy and apoptosis biological processes. Thus, achieving a better understanding of the autophagy and apoptosis processes and the proteins that crosslinked both pathways, could provide new insights for the MM treatment and improve the development of novel therapeutic strategies to overcome resistance. This review presents a sufficient overview of the roles of autophagy and apoptosis and how they crosslink and control MM progression and drug resistance. Potential combination targeting of both pathways for improving outcomes in MM patients also has been addressed.
Collapse
Affiliation(s)
- Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA
| | - Daniel A. Guirguis
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Nicole K. Schmalbach
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Gabriella Yao
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | | | | | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Correspondence: ; Tel.: +1-856-956-2751
| |
Collapse
|
38
|
Long X, Li J, Wen F, Cao Y, Luo Z, Luo C. miR-140-3p attenuated the tumorigenesis of multiple myeloma via attenuating BZW2. Hematology 2022; 27:173-180. [PMID: 35068373 DOI: 10.1080/16078454.2021.2009644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Among B-cell lymphoma, multiple myeloma (MM) is an incurable malignancy. miR-140-3p was known to be an inhibitor in malignant tumors. However, the function of miR-140-3p in MM remains unclear. METHODS qRT-PCR was performed to determine the expressions of miR-140-3p and BZW2 mRNA. The protein level of BZW2 was determined by the western blot. Cell viability or cell apoptosis was detected by the MTT assay or flow cytometry, respectively. Binding between miR-140-3p and BZW2 was validated using the dual luciferase assay. Xenograft model was applied to verify the results of in vitro study. RESULTS The level of miR-140-3p was significantly downregulated in MM. Overpexression of miR-140-3p impaired the proliferation of MM cell lines and induced apoptosis in MM cells. miR-140-3p was validated to target BZW2 and inhibit the expression of BZW2. BZW2 was involved in the regulation of miR-140-3p on MM cell vitality and apoptosis. In vivo study revealed that miR-140-3p impeded tumorigenesis of MM cell line in nude mice. CONCLUSION Our present study revealed that miR-140-3p served as a suppressor in MM by negatively regulating BZW2. Thus, miR-140-3p could act as a new target for treating MM.
Collapse
Affiliation(s)
- Xingxing Long
- Department of Hematology, The 1st Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Junjun Li
- Department of Hematology, The 1st Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Feng Wen
- Department of Hematology, The 1st Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Yixiong Cao
- Department of Hematology, The 1st Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Zeyu Luo
- Department of Hematology, The 1st Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Cong Luo
- Department of Hematology, The 1st Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| |
Collapse
|
39
|
Liu Q, Ran R, Song M, Li X, Wu Z, Dai G, Xia R. LncRNA HCP5 acts as a miR-128-3p sponge to promote the progression of multiple myeloma through activating Wnt/β-catenin/cyclin D1 signaling via PLAGL2. Cell Biol Toxicol 2022; 38:979-993. [PMID: 34331612 DOI: 10.1007/s10565-021-09628-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although long non-coding RNA (lncRNA) HCP plays essential roles in human cancers, its function and mechanism in multiple myeloma (MM) have not crystallized. METHODS HCP5 level in MM was assessed through qRT-PCR. A series of functional investigations were conducted to evaluate the influences of HCP5 on proliferation and apoptosis. Bioinformatics analysis and RIP/RNA pull-down assays were carried out to determine the relationships among HCP5, miR-128-3p, and PLAGL2. Relative protein level was determined through Western blot. A xenograft tumor model was applied for validating the roles of HCP5/miR-128-3p/PLAGL2 axis in vivo. RESULTS HCP5 was significantly increased in MM. HCP5 knockdown effectively thwarted the proliferative rate and cell cycle of MM cell lines and suppressed tumor growth. HCP5 regulated PLAGL2 expression by sponging miR-128-3p. PLAGL2 overexpression effectively rescued cells from influences by sh-HCP5 on cell proliferative and apoptotic rates. Additionally, HCP5 knockdown significantly inhibited Wnt/β-catenin/cyclin D1 signaling, and these effects were eliminated by PLAGL2 overexpression. CONCLUSION Our study revealed that HCP5/miR-128-3p/PLAGL2 is closely correlated to MM development by modulating Wnt/β-catenin/cyclin D1 signaling. HCP5 promoted cell proliferation and tumor formation of MM cells by activating the Wnt/β-catenin/CCND1 signaling pathway by sponging miR-128-3p to increase PLAGL2 expression.
Collapse
Affiliation(s)
- Qinhua Liu
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Ruonan Ran
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingyue Song
- Department of Hematology, the Chaohu Hospital Affiliated to Anhui Medical University, Chaohu, Anhui, China
| | - Xiaodan Li
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Zhengsheng Wu
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guanrong Dai
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Ruixiang Xia
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
| |
Collapse
|
40
|
Xiong Q, Zhang Y, Li J, Zhu Q. Small Non-Coding RNAs in Human Cancer. Genes (Basel) 2022; 13:genes13112072. [PMID: 36360311 PMCID: PMC9690286 DOI: 10.3390/genes13112072] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Small non-coding RNAs are widespread in the biological world and have been extensively explored over the past decades. Their fundamental roles in human health and disease are increasingly appreciated. Furthermore, a growing number of studies have investigated the functions of small non-coding RNAs in cancer initiation and progression. In this review, we provide an overview of the biogenesis of small non-coding RNAs with a focus on microRNAs, PIWI-interacting RNAs, and a new class of tRNA-derived small RNAs. We discuss their biological functions in human cancer and highlight their clinical application as molecular biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Qunli Xiong
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaguang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junjun Li
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qing Zhu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence:
| |
Collapse
|
41
|
MiR-371a-5p Positively Associates with Hepatocellular Carcinoma Malignancy but Sensitizes Cancer Cells to Oxaliplatin by Suppressing BECN1-Dependent Autophagy. Life (Basel) 2022; 12:life12101651. [DOI: 10.3390/life12101651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 01/27/2023] Open
Abstract
Oxaliplatin (OXA)-based chemotherapy demonstrates active efficacy in advanced hepatocellular carcinoma (HCC), while resistance development limits its clinical efficacy. Thus, identifying resistance-related molecules and underlying mechanisms contributes to improving the therapeutic efficacy of HCC patients. MicroRNA-371a-5p (MiR-371a-5p) fulfills an important function in tumor progression. However, little is known about the effect of miR-371a-5p on chemotherapy response. In this study, quantitative real-time polymerase chain reaction, Western blot and immunohistochemistry were used to determine the expression levels of miR-371a-5p, BECN1 and autophagy-related proteins in HCC cells, tissues and serum. The luciferase reporter assay was used to assess the directly suppressive effect of miR-371a-5p on BECN1 mRNA translation. Moreover, gain- and loss-of-function assays and rescue assays were used to evaluate the mediated effect of BECN1-dependent autophagy on the role of miR-371a-5p in the response of HCC cells to OXA. We found that miR-371a-5p was significantly up-regulated in HCC tissues and serum from patients, whereas BECN1 protein was down-regulated in HCC tissues compared to the corresponding controls. We also found that there was a negative correlation between the two molecules in HCC tissues. In addition, we found that miR-371a-5p expression was positively associated with malignant characteristics of HCC and BECN1 protein expression is negatively associated. Contrary to this, we found that miR-371a-5p enhances and BECN1 attenuates the response of HCC cells to OXA. Importantly, the enhanced effect of miR-371a-5p on the response of HCC cells to OXA could be reduced by re-expression of non-targetable BECN1, and then the reduced effect was restored following bafilomycin A treatment. Taken together, we identified a dual role of miR-371a-5p in HCC malignant characteristics and the response of HCC cells to oxaliplatin. Importantly, we reveal that miR-371a-5p enhances oxaliplatin response by target suppression of BECN1-dependent autophagy.
Collapse
|
42
|
Chen Y, Zhang C, Du Y, Yang X, Liu M, Yang W, Lei G, Wang G. Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6. Chin Med J (Engl) 2022; 135:2467-2477. [PMID: 36449688 PMCID: PMC9945297 DOI: 10.1097/cm9.0000000000002377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication in patients, especially elderly patients, who undergo cardiac surgery with cardiopulmonary bypass. Studies have indicated a protective role of autophagy in AKI. However, the mechanisms underlying the regulatory effect of autophagy in AKI among patients undergoing cardiac surgeries are poorly understood. In this study, we aimed to test the hypothesis that exosomal microRNAs (miRNAs) regulate autophagy in tubular epithelial cells after AKI. METHODS Plasma exosomal RNA was extracted from young and elderly AKI patients undergoing cardiac surgery, and the miRNAs expression during the perioperative period were analyzed using next-generation sequencing. The screened miRNAs and their target genes were subjected to gene oncology function and Kyoto Encyclopedia of Genes and Genome enrichment analyses. Renal tubular epithelial cell line (HK-2 cells) was cultured and hypoxia/reoxygenation (H/R) model was established, which is an in vitro renal ischemia/reperfusion (I/R) model. We used Western blot analysis, cell viability assay, transfection, luciferase assay to investigate the mechanisms underlying the observed increases in the levels of renal I/R injury-mediated exosomal miRNAs and their roles in regulating HK-2 cells autophagy. RESULTS miR-590-3p was highly enriched in the plasma exosomes of young AKI patients after cardiac surgery. Increased levels of miR-590-3p led to the increases in the expression of autophagy marker proteins, including Beclin-1 and microtubule associated protein 1 light chain 3 beta (LC3II), and prolonged the autophagic response in HK-2 cells after H/R treatment. These effects were achieved mainly via increases in the exosomal miR-590-3p levels, and the tumor necrosis factor receptor-associated factor 6 protein was shown to play a key role in I/R injury-mediated autophagy induction. CONCLUSION Exosomes released from HK-2 cells after renal I/R injury regulate autophagy by transferring miR-590-3p in a paracrine manner, which suggests that increasing the miR-590-3p levels in HK-2 cell-derived exosomes may increase autophagy and protect against kidney injury after renal I/R injury.
Collapse
Affiliation(s)
- Yimeng Chen
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Congya Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yingjie Du
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiying Yang
- Weifang Medical University, School of Anesthesiology, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang, Shandong 261053, China
| | - Min Liu
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Wenjing Yang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Guiyu Lei
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
43
|
Noncoding RNAs: Regulating the crosstalk between tumor-associated macrophages and gastrointestinal cancer. Biomed Pharmacother 2022; 153:113370. [DOI: 10.1016/j.biopha.2022.113370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/19/2023] Open
|
44
|
Toraih EA, Fawzy MS, Ning B, Zerfaoui M, Errami Y, Ruiz EM, Hussein MH, Haidari M, Bratton M, Tortelote GG, Hilliard S, Nilubol N, Russell JO, Shama MA, El-Dahr SS, Moroz K, Hu T, Kandil E. A miRNA-Based Prognostic Model to Trace Thyroid Cancer Recurrence. Cancers (Basel) 2022; 14:cancers14174128. [PMID: 36077665 PMCID: PMC9454675 DOI: 10.3390/cancers14174128] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/12/2022] [Accepted: 08/21/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Some thyroid tumors elected for surveillance remain indolent, while others progress. The mechanism responsible for this difference is poorly understood, making it challenging to devise patient surveillance plans. Early prediction is important for tailoring treatment and follow-up in high-risk patients. The aim of our study was to identify predictive markers for progression. We leveraged a highly sensitive test that accurately predicts which thyroid nodules are more likely to develop lymph node metastasis, thereby improving care and outcomes for cancer patients. Abstract Papillary thyroid carcinomas (PTCs) account for most endocrine tumors; however, screening and diagnosing the recurrence of PTC remains a clinical challenge. Using microRNA sequencing (miR-seq) to explore miRNA expression profiles in PTC tissues and adjacent normal tissues, we aimed to determine which miRNAs may be associated with PTC recurrence and metastasis. Public databases such as TCGA and GEO were utilized for data sourcing and external validation, respectively, and miR-seq results were validated using quantitative real-time PCR (qRT-PCR). We found miR-145 to be significantly downregulated in tumor tissues and blood. Deregulation was significantly related to clinicopathological features of PTC patients including tumor size, lymph node metastasis, TNM stage, and recurrence. In silico data analysis showed that miR-145 can negatively regulate multiple genes in the TC signaling pathway and was associated with cell apoptosis, proliferation, stem cell differentiation, angiogenesis, and metastasis. Taken together, the current study suggests that miR-145 may be a biomarker for PTC recurrence. Further mechanistic studies are required to uncover its cellular roles in this regard.
Collapse
Affiliation(s)
- Eman A. Toraih
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: ; Tel.: +1-346-907-4237
| | - Manal S. Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar P.O. Box 1321, Saudi Arabia
| | - Bo Ning
- Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Mourad Zerfaoui
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Youssef Errami
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Emmanuelle M. Ruiz
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Mohammad H. Hussein
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Muhib Haidari
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Melyssa Bratton
- Biospecimen Core Laboratory, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Giovane G. Tortelote
- Section of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Sylvia Hilliard
- Section of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Naris Nilubol
- Endocrine Oncology Branch, National Cancer Institute, National Institute of Health, Bethesda, MD 20814, USA
| | - Jonathon O. Russell
- Division of Head and Neck Endocrine Surgery, Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins, Baltimore, MD 21287, USA
| | - Mohamed A. Shama
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Samir S. El-Dahr
- Section of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Krzysztof Moroz
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Tony Hu
- Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Emad Kandil
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
45
|
Implication of microRNAs in Carcinogenesis with Emphasis on Hematological Malignancies and Clinical Translation. Int J Mol Sci 2022; 23:ijms23105838. [PMID: 35628648 PMCID: PMC9143361 DOI: 10.3390/ijms23105838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/30/2022] Open
Abstract
MicroRNAs (miRNAs) are evolutionarily conserved small non-coding RNAs, that are involved in the multistep process of carcinogenesis, contributing to all established hallmarks of cancer. In this review, implications of miRNAs in hematological malignancies and their clinical utilization fields are discussed. As components of the complex regulatory network of gene expression, influenced by the tissue microenvironment and epigenetic modifiers, miRNAs are “micromanagers” of all physiological processes including the regulation of hematopoiesis and metabolic pathways. Dysregulated miRNA expression levels contribute to both the initiation and progression of acute leukemias, the metabolic reprogramming of malignantly transformed hematopoietic precursors, and to the development of chemoresistance. Since they are highly stable and can be easily quantified in body fluids and tissue specimens, miRNAs are promising biomarkers for the early detection of hematological malignancies. Besides novel opportunities for differential diagnosis, miRNAs can contribute to advanced chemoresistance prediction and prognostic stratification of acute leukemias. Synthetic oligonucleotides and delivery vehicles aim the therapeutic modulation of miRNA expression levels. However, major challenges such as efficient delivery to specific locations, differences of miRNA expression patterns between pediatric and adult hematological malignancies, and potential side effects of miRNA-based therapies should be considered.
Collapse
|
46
|
Zhu T, Cen Y, Chen Z, Zhang Y, Zhao L, Wang J, Lu W, Xie X, Wang X. Oncogenic circTICRR suppresses autophagy via binding to HuR protein and stabilizing GLUD1 mRNA in cervical cancer. Cell Death Dis 2022; 13:479. [PMID: 35595754 PMCID: PMC9122915 DOI: 10.1038/s41419-022-04943-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Circular RNAs (circRNAs) are critical regulators in the occurrence and development of numerous cancers, in which abnormal autophagy plays a key role. However, the potential involvement of circRNAs in autophagy is largely unknown. Here, we identified the overexpression of circTICRR, a circular RNA, in cervical cancer. In vitro experiments showed that knockdown of circTICRR activated autophagy, and consequently promoted apoptosis and inhibited proliferation in cervical cancer cells, and vice versa. CircTICRR interacted with HuR protein via binding to F287/F289 in the RRM3 domain of HuR, stabilizing GLUD1 mRNA and elevating the level of GLUD1 protein. In vivo experiments revealed that knockdown of circTICRR suppressed the growth of transplanted tumors. An inhibitory peptide specific to the binding site between circTICRR and HuR protein promoted autophagy, induced apoptosis, suppressed proliferation in cervical cancer cells, and inhibited the growth of xenografts. Our findings suggest that circTICRR acts as an oncogene in cervical cancer and the interaction between circTICRR and HuR protein may be a potential target in cervical cancer therapeutics.
Collapse
Affiliation(s)
- Tingjia Zhu
- grid.13402.340000 0004 1759 700XWomen’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China
| | - Yixuan Cen
- grid.13402.340000 0004 1759 700XWomen’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China
| | - Zhuoye Chen
- grid.13402.340000 0004 1759 700XWomen’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China
| | - Yanan Zhang
- grid.13402.340000 0004 1759 700XDepartment of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China
| | - Lu Zhao
- grid.13402.340000 0004 1759 700XDepartment of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China
| | - Jiaying Wang
- grid.414252.40000 0004 1761 8894Department of Obstetrics and Gynecology, The Seventh Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Weiguo Lu
- grid.13402.340000 0004 1759 700XWomen’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China ,grid.13402.340000 0004 1759 700XDepartment of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer center, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Xing Xie
- grid.13402.340000 0004 1759 700XWomen’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China ,grid.13402.340000 0004 1759 700XDepartment of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China
| | - Xinyu Wang
- grid.13402.340000 0004 1759 700XWomen’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China ,grid.13402.340000 0004 1759 700XDepartment of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer center, Zhejiang University, Hangzhou, 310058 Zhejiang China ,grid.13402.340000 0004 1759 700XDepartment of Gynecology and Obstetrics, The First Affiliated Hospital School of Medicine, Zhejiang University, Hangzhou, 310003 Zhejiang China
| |
Collapse
|
47
|
Mahabady MK, Mirzaei S, Saebfar H, Gholami MH, Zabolian A, Hushmandi K, Hashemi F, Tajik F, Hashemi M, Kumar AP, Aref AR, Zarrabi A, Khan H, Hamblin MR, Nuri Ertas Y, Samarghandian S. Noncoding RNAs and their therapeutics in paclitaxel chemotherapy: Mechanisms of initiation, progression, and drug sensitivity. J Cell Physiol 2022; 237:2309-2344. [PMID: 35437787 DOI: 10.1002/jcp.30751] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
The identification of agents that can reverse drug resistance in cancer chemotherapy, and enhance the overall efficacy is of great interest. Paclitaxel (PTX) belongs to taxane family that exerts an antitumor effect by stabilizing microtubules and inhibiting cell cycle progression. However, PTX resistance often develops in tumors due to the overexpression of drug transporters and tumor-promoting pathways. Noncoding RNAs (ncRNAs) are modulators of many processes in cancer cells, such as apoptosis, migration, differentiation, and angiogenesis. In the present study, we summarize the effects of ncRNAs on PTX chemotherapy. MicroRNAs (miRNAs) can have opposite effects on PTX resistance (stimulation or inhibition) via influencing YES1, SK2, MRP1, and STAT3. Moreover, miRNAs modulate the growth and migration rates of tumor cells in regulating PTX efficacy. PIWI-interacting RNAs, small interfering RNAs, and short-hairpin RNAs are other members of ncRNAs regulating PTX sensitivity of cancer cells. Long noncoding RNAs (LncRNAs) are similar to miRNAs and can modulate PTX resistance/sensitivity by their influence on miRNAs and drug efflux transport. The cytotoxicity of PTX against tumor cells can also be affected by circular RNAs (circRNAs) and limitation is that oncogenic circRNAs have been emphasized and experiments should also focus on onco-suppressor circRNAs.
Collapse
Affiliation(s)
- Mahmood K Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Hamidreza Saebfar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad H Gholami
- Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Amirhossein Zabolian
- Resident of Orthopedics, Department of Orthopedics, School of Medicine, 5th Azar Hospital, Golestan University of Medical Sciences, Golestan, Iran
| | - Kiavash Hushmandi
- Division of Epidemiology, Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alan P Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amir R Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.,Xsphera Biosciences Inc, Boston, Massachusetts, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul, Turkey
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey.,ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
48
|
HDACs and the epigenetic plasticity of cancer cells: Target the complexity. Pharmacol Ther 2022; 238:108190. [PMID: 35430294 DOI: 10.1016/j.pharmthera.2022.108190] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022]
Abstract
Cancer cells must adapt to the hostile conditions of the microenvironment in terms of nutrition, space, and immune system attack. Mutations of DNA are the drivers of the tumorigenic process, but mutations must be able to hijack cellular functions to sustain the spread of mutant genomes. Transcriptional control is a key function in this context and is controlled by the rearrangement of the epigenome. Unlike genomic mutations, the epigenome of cancer cells can in principle be reversed. The discovery of the first epigenetic drugs triggered a contaminating enthusiasm. Unfortunately, the complexity of the epigenetic machinery has frustrated this enthusiasm. To develop efficient patient-oriented epigenetic therapies, we need to better understand the nature of this complexity. In this review, we will discuss recent advances in understanding the contribution of HDACs to the maintenance of the transformed state and the rational for their selective targeting.
Collapse
|
49
|
Zhang T, Wang LL, Guan J, Zhou Y, Cheng P, Zou L. MicroRNA-125a/b-5p promotes malignant behavior in multiple myeloma cells and xenograft tumor growth by targeting DIS3. Kaohsiung J Med Sci 2022; 38:574-584. [PMID: 35394705 DOI: 10.1002/kjm2.12534] [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: 09/26/2021] [Revised: 12/17/2021] [Accepted: 01/26/2022] [Indexed: 11/11/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy with a high prevalence and is characterized by the clonal expansion of malignant plasma cells. As a new tumor suppressor, defective in sister chromatid joining (DIS3) was reported to be a gene closely related to MM. This study elucidated the biological functions and underlying mechanisms of DIS3 in MM. DIS3 mRNA and protein levels were detected using RT-qPCR and western blotting, respectively. Methyl thiazolyl tetrazolium assays, flow cytometry analyses, Transwell assays, and wound healing assays were performed to detect the proliferation, apoptosis, invasion, and migration of MM cells. The binding relationship between miR-125a/b-5p and DIS3 was verified using luciferase reporter assays and RNA pulldown assays. Xenograft tumor models were established in nude mice to investigate the effects of miR-125a/b-5p and DIS3 on tumor growth in vivo. DIS3 levels were downregulated in MM cells, and DIS3 upregulation inhibited the malignant behaviors of MM cells. Mechanistically, miR-125a/b-5p directly targeted the 3' untranslated region of DIS3. The expression of miR-125a/b-5p was upregulated in MM cells, miR-125a/b-5p knockdown inhibited the malignant behaviors of MM cells, and the inhibitory effect was reversed by DIS3 downregulation. The results of in vivo experiments indicated that miR-125a/b-5p promoted tumor growth by downregulating DIS3. Overall, miR-125a/b-5p promotes MM cellular processes and xenograft tumor growth by targeting DIS3.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Lan-Lan Wang
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Jun Guan
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Ying Zhou
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Ping Cheng
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Liang Zou
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| |
Collapse
|
50
|
de la Cruz-Ojeda P, Flores-Campos R, Navarro-Villarán E, Muntané J. The Role of Non-Coding RNAs in Autophagy During Carcinogenesis. Front Cell Dev Biol 2022; 10:799392. [PMID: 35309939 PMCID: PMC8926078 DOI: 10.3389/fcell.2022.799392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Macroautophagy (autophagy herein) is a cellular stress response and a survival pathway involved in self-renewal and quality control processes to maintain cellular homeostasis. The alteration of autophagy has been implicated in numerous diseases such as cancer where it plays a dual role. Autophagy serves as a tumor suppressor in the early phases of cancer formation with the restoration of homeostasis and eliminating cellular altered constituents, yet in later phases, autophagy may support and/or facilitate tumor growth, metastasis and may contribute to treatment resistance. Key components of autophagy interact with either pro- and anti-apoptotic factors regulating the proximity of tumor cells to apoptotic cliff promoting cell survival. Autophagy is regulated by key cell signaling pathways such as Akt (protein kinase B, PKB), mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) involved in cell survival and metabolism. The expression of critical members of upstream cell signaling, as well as those directly involved in the autophagic and apoptotic machineries are regulated by microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). Consequently, non-coding RNAs play a relevant role in carcinogenesis and treatment response in cancer. The review is an update of the current knowledge in the regulation by miRNA and lncRNA of the autophagic components and their functional impact to provide an integrated and comprehensive regulatory network of autophagy in cancer.
Collapse
Affiliation(s)
- Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| | - Rocío Flores-Campos
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|