1
|
Wen X, Cheng B, Tan M, Zhang X, Yao Z, Huang R, Zhang G, Liu L, Cai F, Liu H. Investigation of Substance Basis and Pharmacodynamics of Qingzao Jiufei Decoction in Lung Cancer Based On Serum Pharmacochemistry and Proteomics. Chem Biodivers 2025:e202402774. [PMID: 39888178 DOI: 10.1002/cbdv.202402774] [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: 10/28/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/01/2025]
Abstract
Qingzao Jiufei Decoction (QD) is a renowned prescription for nourishing lung Yin to treat lung-related diseases in ancient and modern times. It has been reported that QD can effectively inhibit the growth of lung cancer (LC), but the material basis and mechanism of its treatment of LC remain to be further studied. In this experiment, we combined serum pharmacochemistry and proteomics to study the pharmacodynamic material basis and mechanism of QD against LC. The results showed that in vitro a total of 121 compounds were identified. A total of 33 prototype chemical components were identified in vivo, which may be effective active ingredients for QD treatment of LC. Proteomic experiments have shown that compared with the model group, 113 DEPs in LC tissues were significantly changed after QD treatment, among which 57 proteins were significantly up-regulated and 56 proteins were significantly down-regulated (p < 0.05). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was performed on these DEPs. A total of 205 pathways were enriched, and the PI3K-Akt signaling pathway was experimentally verified. The results showed that QD can be used to treat LC by inhibiting the activation of the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaoli Wen
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Biting Cheng
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Min Tan
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Xiang Zhang
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Ziheng Yao
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Rumei Huang
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Ge Zhang
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Ling Liu
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| | - Fangyan Cai
- Nanchang Medical College, Nanchang, Jiangxi, China
| | - Hongning Liu
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Research Center for Differentiation and Development of CM Basic Theory of Jiangxi University of Chinese Medicine, Jiangxi Province Key Laboratory of CM Etiopathogenesis, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Korns J, Wicker CA, Lehn M, Shyamsunder S, Thompson S, Lester C, Wise-Draper TM, Waltz SE, Takiar V. Telaglenastat as an alternative to cisplatin as a radiosensitizer in the treatment of head and neck squamous cell carcinoma. Cancer Lett 2024; 606:217320. [PMID: 39489210 PMCID: PMC11583984 DOI: 10.1016/j.canlet.2024.217320] [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: 04/30/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
The efficacy of radiation treatment (RT) of head and neck squamous cell carcinoma (HNSCC) is limited by radioresistance and the toxicity of FDA approved radiosensitizers. In extension to our previous research where we demonstrated that telaglenastat (CB839) increased efficacy of RT in in vitro and in vivo HNSCC models, here, we examine the radiosensitizing effects of telaglenastat in comparison to cisplatin's, as cisplatin is currently the standard of care for concurrent therapy. Combination of telaglenastat with RT reduced tumor volume in a HNSCC patient derived xenograft mouse model. The efficacy of telaglenastat with RT in reducing cell survival and increasing apoptosis was similar if not greater than that of cisplatin with RT in Cal27 and HN5 HNSCC cells. The addition of telaglenastat increased reactive oxygen species and reduced the antioxidant glutathione in both Cal27 and HN5 cells. Reverse Phase Protein Array analyses revealed alterations in cell death and DNA damage response proteins. This study provides the scientific underpinnings for the use of telaglenastat as a radiosensitizer in the treatment of HNSCC either as an alternative to cisplatin or in cisplatin-ineligible patients.
Collapse
Affiliation(s)
- Julianna Korns
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Christina A Wicker
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Maria Lehn
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Shreya Shyamsunder
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Samuel Thompson
- Cincinnati Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Carissa Lester
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Trisha M Wise-Draper
- Department of Internal Medicine, Division of Hematology Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA; Research Service, Cincinnati Veteran's Affairs Medical Center, Cincinnati, OH, USA
| | - Vinita Takiar
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA; Research Service, Cincinnati Veteran's Affairs Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
3
|
Lara P, Quiñonero F, Ortiz R, Prados J, Melguizo C. Nanoparticles Bounded to Interfering RNAs as a Therapy for Pancreatic Cancer: A Systematic Review. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2013. [PMID: 39510122 DOI: 10.1002/wnan.2013] [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: 04/30/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Pancreatic cancer is one of the tumors with poor prognosis and low survival due to late diagnosis, high resistance, and very limited effective therapeutic options. Thus, new pharmacological treatments are necessary to improve the prognosis of patients. In this context, nanoparticles represent an efficient system for transporting and administering therapeutic molecules. Furthermore, siRNA can be used in cancer treatment to selectively inhibit the expression of any target gene. Therefore, nanoparticles associated with siRNA have been tested as a new therapeutic strategy to solve the pancreatic cancer treatment failure in the clinical setting. The current article presents a systematic revision of the literature of the last 10 years in which nanoparticles loading siRNA are used in pancreatic cancer. This research was carried out in three databases (PubMed, Scopus, and Web of Science) obtaining 164 articles from which 37 were selected. Our results show an overall view of the high effectiveness of this new therapy that combines nanoparticles with genetic therapy in pancreatic cancer suggesting that siRNA-based medicines will likely open up a new therapeutic era in the treatment of this type of tumors.
Collapse
Affiliation(s)
- Patricia Lara
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
| | - Francisco Quiñonero
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
| | - Raul Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (Ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada, Spain
| |
Collapse
|
4
|
Korzun T, Moses AS, Diba P, Sattler AL, Olson B, Taratula OR, Pejovic T, Marks DL, Taratula O. Development and Perspectives: Multifunctional Nucleic Acid Nanomedicines for Treatment of Gynecological Cancers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301776. [PMID: 37518857 PMCID: PMC10827528 DOI: 10.1002/smll.202301776] [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: 02/28/2023] [Revised: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Gynecological malignancies are a significant cause of morbidity and mortality across the globe. Due to delayed presentation, gynecological cancer patients are often referred late in the disease's course, resulting in poor outcomes. A considerable number of patients ultimately succumb to chemotherapy-resistant disease, which reoccurs at advanced stages despite treatment interventions. Although efforts have been devoted to developing therapies that demonstrate reduced resistance to chemotherapy and enhanced toxicity profiles, current clinical outcomes remain unsatisfactory due to treatment resistance and unfavorable off-target effects. Consequently, innovative biological and nanotherapeutic approaches are imperative to strengthen and optimize the therapeutic arsenal for gynecological cancers. Advancements in nanotechnology-based therapies for gynecological malignancies offer significant advantages, including reduced toxicity, expanded drug circulation, and optimized therapeutic dosing, ultimately leading to enhanced treatment effectiveness. Recent advances in nucleic acid therapeutics using microRNA, small interfering RNA, and messenger RNA provide novel approaches for cancer therapeutics. Effective single-agent and combinatorial nucleic acid therapeutics for gynecological malignancies have the potential to transform cancer treatment by giving safer, more tailored approaches than conventional therapies. This review highlights current preclinical studies that effectively exploit these approaches for the treatment of gynecological malignant tumors and malignant ascites.
Collapse
Affiliation(s)
- Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue Portland, Portland, OR, 97239, USA
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
| | - Ariana L Sattler
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, Oregon, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Brennan Olson
- Mayo Clinic Department of Otolaryngology-Head and Neck Surgery, 200 First St. SW, Rochester, MN, 55905, USA
| | - Olena R Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Tanja Pejovic
- Departments of Obstetrics and Gynecology and Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, Oregon, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue Portland, Portland, OR, 97239, USA
| |
Collapse
|
5
|
Maffini F, Lepanto D, Chu F, Tagliabue M, Vacirca D, De Berardinis R, Gandini S, Vignati S, Ranghiero A, Taormina S, Rappa A, Cossu Rocca M, Alterio D, Chiocca S, Barberis M, Preda L, Pagni F, Fusco N, Ansarin M. A Transcriptomic Analysis of Laryngeal Dysplasia. Int J Mol Sci 2024; 25:9685. [PMID: 39273632 PMCID: PMC11395940 DOI: 10.3390/ijms25179685] [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: 07/31/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
This article describes how the transcriptional alterations of the innate immune system divide dysplasias into aggressive forms that, despite the treatment, relapse quickly and more easily, and others where the progression is slow and more treatable. It elaborates on how the immune system can change the extracellular matrix, favoring neoplastic progression, and how infections can enhance disease progression by increasing epithelial damage due to the loss of surface immunoglobulin and amplifying the inflammatory response. We investigated whether these dysregulated genes were linked to disease progression, delay, or recovery. These transcriptional alterations were observed using the RNA-based next-generation sequencing (NGS) panel Oncomine Immune Response Research Assay (OIRRA) to measure the expression of genes associated with lymphocyte regulation, cytokine signaling, lymphocyte markers, and checkpoint pathways. During the analysis, it became apparent that certain alterations divide dysplasia into two categories: progressive or not. In the future, these biological alterations are the first step to provide new treatment modalities with different classes of drugs currently in use in a systemic or local approach, including classical chemotherapy drugs such as cisplatin and fluorouracile, older drugs like fenretinide, and new checkpoint inhibitor drugs such as nivolumab and pembrolizumab, as well as newer options like T cell therapy (CAR-T). Following these observed alterations, it is possible to differentiate which dysplasias progress or not or relapse quickly. This information could, in the future, be the basis for determining a close follow-up, minimizing surgical interventions, planning a correct and personalized treatment protocol for each patient and, after specific clinical trials, tailoring new drug treatments.
Collapse
Affiliation(s)
- Fausto Maffini
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Daniela Lepanto
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesco Chu
- Division of Otolaryngology Head and Neck Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Marta Tagliabue
- Division of Otolaryngology Head and Neck Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Davide Vacirca
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Rita De Berardinis
- Division of Otolaryngology Head and Neck Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Sara Gandini
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Silvano Vignati
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Alberto Ranghiero
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Sergio Taormina
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Alessandra Rappa
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Maria Cossu Rocca
- Medical Oncology Division of Urogenital and Head and Neck Tumors, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Daniela Alterio
- Department of Radiotherapy, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Susanna Chiocca
- Department of Experimental Oncology, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Massimo Barberis
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenzo Preda
- Diagnostic Imaging Unit, National Center of Oncological Hadron-Therapy (CNAO), 27100 Pavia, Italy;
- State University School of Medicine, University of Pavia, 27100 Pavia, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo dei Tintori, University of Milano-Bicocca, 20126 Milan, Italy
| | - Nicola Fusco
- Department of Surgical Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
- State University School of Medicine, University of Milan, 20122 Milan, Italy
| | - Mohssen Ansarin
- Division of Otolaryngology Head and Neck Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
6
|
Chen B, Liu J. Advances in ovarian tumor stem cells and therapy. Cell Biochem Biophys 2024; 82:1871-1892. [PMID: 38955927 DOI: 10.1007/s12013-024-01385-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Ovarian cancer is considered the most lethal among all gynecological malignancies due to its early metastatic dissemination, extensive spread, and malignant ascites. The current standard of care for advanced ovarian cancer involves a combination of cytoreductive surgery and chemotherapy utilizing platinum-based and taxane-based agents. Although initial treatment yields clinical remission in 70-80% of patients, the majority eventually develop treatment resistance and tumor recurrence. A growing body of evidence indicates the existence of cancer stem cells within diverse solid tumors, including ovarian cancer, which function as a subpopulation to propel tumor growth and disease advancement by means of drug resistance, recurrence, and metastasis. The presence of ovarian cancer stem cells is widely considered to be a significant contributor to the unfavorable clinical outcomes observed in patients with ovarian cancer, as they play a crucial role in mediating chemotherapy resistance, recurrence, and metastasis. Ovarian cancer stem cells possess the capacity to reassemble within the entirety of the tumor following conventional treatment, thereby instigating the recurrence of ovarian cancer and inducing resistance to treatment. Consequently, the creation of therapeutic approaches aimed at eliminating ovarian cancer stem cells holds great potential for the management of ovarian cancer. These cells are regarded as one of the most auspicious targets and mechanisms for the treatment of ovarian cancer. There is a pressing need for a comprehensive comprehension of the fundamental mechanisms of ovarian cancer's recurrence, metastasis, and drug resistance, alongside the development of effective strategies to overcome chemoresistance, metastasis, and recurrence. The implementation of cancer stem cell therapies may potentially augment the tumor cells' sensitivity to existing chemotherapy protocols, thereby mitigating the risks of tumor metastasis and recurrence, and ultimately improving the survival rates of ovarian cancer patients.
Collapse
Affiliation(s)
- Biqing Chen
- Harbin Medical University, Harbin, Heilongjiang, China.
| | - Jiaqi Liu
- Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
7
|
Chawhan AP, Dsouza N. Identifying the key hub genes linked with lung squamous cell carcinoma by examining the differentially expressed and survival genes. Mol Genet Genomics 2024; 299:76. [PMID: 39097557 DOI: 10.1007/s00438-024-02169-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Lung Squamous Cell Carcinoma is characterised by significant alterations in RNA expression patterns, and a lack of early symptoms and diagnosis results in poor survival rates. Our study aimed to identify the hub genes involved in LUSC by differential expression analysis and their influence on overall survival rates in patients. Thus, identifying genes with the potential to serve as biomarkers and therapeutic targets. RNA sequence data for LUSC was obtained from TCGA and analysed using R Studio. Survival analysis was performed on DE genes. PPI network and hub gene analysis was performed on survival-relevant genes. Enrichment analysis was conducted on the PPI network to elucidate the functional roles of hub genes. Our analysis identified 2774 DEGs in LUSC patient datasets. Survival analysis revealed 511 genes with a significant impact on patient survival. Among these, 20 hub genes-FN1, ACTB, HGF, PDGFRB, PTEN, SNAI1, TGFBR1, ESR1, SERPINE1, THBS1, PDGFRA, VWF, BMP2, LEP, VTN, PXN, ABL1, ITGA3 and ANXA5-were found to have lower expression levels associated with better patient survival, whereas high expression of SOX2 correlated with longer survival. Enrichment analysis indicated that these hub genes are involved in critical cellular and cancer-related pathways. Our study has identified six key hub genes that are differentially expressed and exhibit significant influence over LUSC patient survival outcomes. Further, in vitro and in vivo studies must be conducted on the key genes for their utilisation as therapeutic targets and biomarkers in LUSC.
Collapse
Affiliation(s)
| | - Norine Dsouza
- Department of Biotechnology, St. Xavier's College, Mumbai, Maharashtra, 400001, India.
| |
Collapse
|
8
|
Wang F, Zhou C, Zhu Y, Keshavarzi M. The microRNA Let-7 and its exosomal form: Epigenetic regulators of gynecological cancers. Cell Biol Toxicol 2024; 40:42. [PMID: 38836981 PMCID: PMC11153289 DOI: 10.1007/s10565-024-09884-3] [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: 01/31/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
Many types of gynecological cancer (GC) are often silent until they reach an advanced stage, and are therefore often diagnosed too late for effective treatment. Hence, there is a real need for more efficient diagnosis and treatment for patients with GC. During recent years, researchers have increasingly studied the impact of microRNAs cancer development, leading to a number of applications in detection and treatment. MicroRNAs are a particular group of tiny RNA molecules that regulate regular gene expression by affecting the translation process. The downregulation of numerous miRNAs has been observed in human malignancies. Let-7 is an example of a miRNA that controls cellular processes as well as signaling cascades to affect post-transcriptional gene expression. Recent research supports the hypothesis that enhancing let-7 expression in those cancers where it is downregulated may be a potential treatment option. Exosomes are tiny vesicles that move through body fluids and can include components like miRNAs (including let-7) that are important for communication between cells. Studies proved that exosomes are able to enhance tumor growth, angiogenesis, chemoresistance, metastasis, and immune evasion, thus suggesting their importance in GC management.
Collapse
Affiliation(s)
- Fei Wang
- Haiyan People's Hospital, Zhejiang Province, Jiaxing, 314300, Zhejiang, China
| | - Chundi Zhou
- Haiyan People's Hospital, Zhejiang Province, Jiaxing, 314300, Zhejiang, China
| | - Yanping Zhu
- Haiyan People's Hospital, Zhejiang Province, Jiaxing, 314300, Zhejiang, China.
| | - Maryam Keshavarzi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Tehran, Iran.
| |
Collapse
|
9
|
Popov A, Hrudka J, Szabó A, Oliverius M, Šubrt Z, Vránová J, Ciprová V, Moravcová J, Mandys V. Expression of Selected miRNAs in Undifferentiated Carcinoma with Osteoclast-like Giant Cells (UCOGC) of the Pancreas: Comparison with Poorly Differentiated Pancreatic Ductal Adenocarcinoma. Biomedicines 2024; 12:962. [PMID: 38790924 PMCID: PMC11117927 DOI: 10.3390/biomedicines12050962] [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: 02/23/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Undifferentiated carcinoma with osteoclast-like giant cells (UCOGC) of the pancreas represents a rare subtype of pancreatic ductal adenocarcinoma (PDAC). Despite a distinct morphology and specific clinical behavior, UCOGCs exhibit unexpected similarities in regard to DNA mutational profiles with conventional PDAC. Treating pancreatic ductal adenocarcinoma is particularly challenging, with limited prospects for cure. As with many other malignant neoplasms, the exploration of microRNAs (miRNAs, miRs) in regulating the biological characteristics of pancreatic cancer is undergoing extensive investigation to enhance tumor diagnostics and unveil the therapeutic possibilities. Herein, we evaluated the expression of miR-21, -96, -148a, -155, -196a, -210, and -217 in UCOGCs and poorly differentiated (grade 3, G3) PDACs. The expression of miR-21, miR-155, and miR-210 in both UCOGCs and G3 PDACs was significantly upregulated compared to the levels in normal tissue, while the levels of miR-148a and miR-217 were downregulated. We did not find any significant differences between cancerous and normal tissues for the expression of miR-96 and miR-196a in G3 PDACs, whereas miR-196a was slightly, but significantly, downregulated in UCOGCs. On the other hand, we have not observed significant differences in the expression of the majority of miRNAs between UCOGC and G3 PDAC, with the exception of miR-155. UCOGC samples demonstrated lower mean levels of miR-155 in comparison with those in G3 PDACs.
Collapse
Affiliation(s)
- Alexey Popov
- Department of Pathology, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic; (A.P.); (A.S.)
| | - Jan Hrudka
- Department of Pathology, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic; (A.P.); (A.S.)
| | - Arpád Szabó
- Department of Pathology, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic; (A.P.); (A.S.)
| | - Martin Oliverius
- Department of Surgery, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic; (M.O.); (Z.Š.)
| | - Zdeněk Šubrt
- Department of Surgery, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic; (M.O.); (Z.Š.)
| | - Jana Vránová
- Department of Medical Biophysics and Medical Informatics, 3rd Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| | - Vanda Ciprová
- Institute of Pathology, 1st Faculty of Medicine, Charles University, General University Hospital, 100 00 Prague, Czech Republic
| | - Jana Moravcová
- Department of Pathology, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic; (A.P.); (A.S.)
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, 140 00 Prague, Czech Republic
| | - Václav Mandys
- Department of Pathology, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic; (A.P.); (A.S.)
| |
Collapse
|
10
|
Xu LJ, Yang Y, Yuan LF, Liu H, Xu NP, Yang Y, Huang L. SP1-stimulated miR-208a-5p aggravates sepsis-induced myocardial injury via targeting XIAP. Exp Cell Res 2024; 435:113905. [PMID: 38163563 DOI: 10.1016/j.yexcr.2023.113905] [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: 09/07/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/03/2024]
Abstract
The development of sepsis can lead to many organ dysfunction and even death. Myocardial injury is one of the serious complications of sepsis leading to death. New evidence suggests that microRNAs (miRNAs) play a critical role in infection myocardial injury. However, the mechanism which miR-208a-5p regulates sepsis-induced myocardial injury remains unclear. To mimic sepsis-induced myocardial injury in vitro, rat primary cardiomyocytes were treated with LPS. Cell viability and apoptosis were tested by CCK-8 and flow cytometry, respectively. The secretion of inflammatory factors was analyzed by ELISA. mRNA and protein levels were detected by RT-qPCR and Western blotting. The interaction among SP1, XIAP and miR-208a-5p was detected using dual luciferase report assay. Ultrasonic analysis and HE staining was performed to observe the effect of miR-208a-5p in sepsis-induced rats. Our findings indicated that miR-208a-5p expression in primary rat cardiomyocytes was increased by LPS. MiR-208a-5p inhibitor reversed LPS-induced cardiomyocytes injury through inhibiting the apoptosis. Furthermore, the inflammatory injury in cardiomyocytes was induced by LPS, which was rescued by miR-208a-5p inhibitor. In addition, downregulation of miR-208a-5p improved LPS-induced sepsis myocardial injury in vivo. Mechanistically, XIAP might be a target gene of miR-208a-5p. SP1 promoted transcription of miR-208a by binding to the miR-208a promoter region. Moreover, silencing of XIAP reversed the regulatory of miR-208a-5p inhibitor on cardiomyocytes injury. To sum up, those findings revealed silencing of miR-208a-5p could alleviate sepsis-induced myocardial injury, which would grant a new process for the treatment of sepsis.
Collapse
Affiliation(s)
- Ling-Jun Xu
- Department of Emergency, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Emergency, Jiangxi Provincial Children's Hospital, Nanchang 330038, Jiangxi Province, PR China
| | - Yixian Yang
- Department of Emergency, Jiangxi Provincial Children's Hospital, Nanchang 330038, Jiangxi Province, PR China
| | - Ling-Feng Yuan
- Department of Function, Jiangxi Provincial Children's Hospital, Nanchang 330038, Jiangxi Province, PR China
| | - Hong Liu
- Department of Emergency, Jiangxi Provincial Children's Hospital, Nanchang 330038, Jiangxi Province, PR China
| | - Nan-Ping Xu
- Department of Emergency, Jiangxi Provincial Children's Hospital, Nanchang 330038, Jiangxi Province, PR China
| | - Yu Yang
- Department of Endocrinology, Metabolism and Genetics, Jiangxi Provincial Children's Hospital, Nanchang 330038, Jiangxi Province, PR China.
| | - Liang Huang
- Department of Emergency, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China.
| |
Collapse
|
11
|
Ajmeera D, Ajumeera R. Drug repurposing: A novel strategy to target cancer stem cells and therapeutic resistance. Genes Dis 2024; 11:148-175. [PMID: 37588226 PMCID: PMC10425757 DOI: 10.1016/j.gendis.2022.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/21/2022] [Accepted: 12/08/2022] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy is an effortless and frequently used approach in cancer therapy. However, in most cases, it can only prolong life expectancy and does not guarantee a complete cure. Furthermore, chemotherapy is associated with severe adverse effects, one of the major complications of effective cancer therapy. In addition, newly published research outputs show that cancer stem cells are involved in cancer disease progression, drug resistance, metastasis, and recurrence and that they are functional in the trans-differentiation capacity of cancer stem cells to cancer cells in response to treatments. Novel strategies are therefore required for better management of cancer therapy. The prime approach would be to synthesize and develop novel drugs that need extensive resources, time, and endurance to be brought into therapeutic use. The subsequent approach would be to screen the anti-cancer activity of available non-cancerous drugs. This concept of repurposing non-cancer drugs as an alternative to current cancer therapy has become popular in recent years because using existing anticancer drugs has several adverse effects. Micronutrients have also been investigated for cancer therapy due to their significant anti-cancer effects with negligible or no side effects and availability in food sources. In this paper, we discuss an ideal hypothesis for screening available non-cancerous drugs with anticancer activity, with a focus on cancer stem cells and their clinical application for cancer treatment. Further, drug repurposing and the combination of micronutrients that can target both cancers and cancer stem cells may result in a better therapeutic approach leading to maximum tumor growth control.
Collapse
Affiliation(s)
- Divya Ajmeera
- Cell Biology Department, ICMR-National Institute of Nutrition (NIN), Hyderabad, Telangana 500007, India
| | - Rajanna Ajumeera
- Cell Biology Department, ICMR-National Institute of Nutrition (NIN), Hyderabad, Telangana 500007, India
| |
Collapse
|
12
|
Suzuki T, Conant A, Curow C, Alexander A, Ioffe Y, Unternaehrer JJ. Role of epithelial-mesenchymal transition factor SNAI1 and its targets in ovarian cancer aggressiveness. JOURNAL OF CANCER METASTASIS AND TREATMENT 2023; 9:25. [PMID: 38009093 PMCID: PMC10673625 DOI: 10.20517/2394-4722.2023.34] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
Ovarian cancer remains the most lethal gynecologic malignancy in the USA. For over twenty years, epithelial-mesenchymal transition (EMT) has been characterized extensively in development and disease. The dysregulation of this process in cancer has been identified as a mechanism by which epithelial tumors become more aggressive, allowing them to survive and invade distant tissues. This occurs in part due to the increased expression of the EMT transcription factor, SNAI1 (Snail). In the case of epithelial ovarian cancer, Snail has been shown to contribute to cancer invasion, stemness, chemoresistance, and metabolic changes. Thus, in this review, we focus on summarizing current findings on the role of EMT (specifically, factors downstream of Snail) in determining ovarian cancer aggressiveness.
Collapse
Affiliation(s)
- Tise Suzuki
- Division of Biochemistry, Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA
| | - Ashlyn Conant
- Division of Biochemistry, Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA
| | - Casey Curow
- Division of Biochemistry, Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA
- University of Redlands, Department of Biology, Redlands, CA 92373, USA
| | - Audrey Alexander
- Division of Biochemistry, Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Natural and Mathematical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
| | - Yevgeniya Ioffe
- Department of Gynecology and Obstetrics, Division of Gynecologic Oncology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Juli J Unternaehrer
- Division of Biochemistry, Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Gynecology and Obstetrics, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
13
|
Chen J, Liu K, Liu W, Yeung WSB. The involvement of let-7 in hCG-induced progesterone synthesis via regulating p27 Kip1 and p21 Cip1 expression. Mol Cell Endocrinol 2023:111970. [PMID: 37230221 DOI: 10.1016/j.mce.2023.111970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 05/27/2023]
Abstract
Progesterone is essential in females to maintain a regular menstrual cycle and pregnancy. The luteinizing hormone (LH) surge induces the luteinization of granulosa cells and thecal cells to form the corpus luteum, which is responsible for progesterone synthesis. However, the specific mechanism of how hCG, the analog of LH, regulates progesterone synthesis has yet to be fully discovered. In this study, we found that progesterone level was increased in adult wild-type pregnant mice 2 and 7 days post-coitum, along with a decrease in let-7 expression compared with the estrus stage. Besides, the let-7 expression was negatively correlated with progesterone level in post-delivery day 23 wild-type female mice after being injected with PMSG and hCG. Then, using let-7 transgenic mice and a human granulosa cell line, we found that overexpression of let-7 antagonized progesterone level via targeting p27Kip1 and p21Cip1 and steroidogenic acute regulatory protein (StAR) expression, which is a rate-limiting enzyme in progesterone synthesis. Furthermore, hCG suppressed let-7 expression by stimulating the MAPK pathway. This study elucidated the role of microRNA let-7 in regulating hCG-induced progesterone production and provided new insights into its role in clinical application.
Collapse
Affiliation(s)
- Jing Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Department of Obstetrics and Gynaecology, Li Kai Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Kui Liu
- Department of Obstetrics and Gynaecology, Li Kai Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Weimin Liu
- Department of Obstetrics and Gynaecology, Li Kai Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
| | - William Shu-Biu Yeung
- Department of Obstetrics and Gynaecology, Li Kai Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
| |
Collapse
|
14
|
Chu F, Maffini F, Lepanto D, Vacirca D, Taormina SV, De Berardinis R, Gandini S, Vignati S, Ranghiero A, Rappa A, Chiocca S, Barberis M, Tagliabue M, Ansarin M. The Genetic and Immunologic Landscape Underlying the Risk of Malignant Progression in Laryngeal Dysplasia. Cancers (Basel) 2023; 15:cancers15041117. [PMID: 36831458 PMCID: PMC9954731 DOI: 10.3390/cancers15041117] [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: 12/27/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
(1) Background: The development of laryngeal cancer is a multistep process involving structural alterations of the epithelial mucosa, from dysplasia (LDy) to invasive carcinoma. In this study, we define new biomarkers, prognostic for malignant transformation, in patients affected by LDy. (2) Methods: We used targeted next-generation sequencing and immunohistochemical analysis to define the mutational and immunological landscape of 15 laryngeal dysplasia progressing to invasive cancer (progressing dysplasia), as well as 31 cases of laryngeal dysplasia that did not progress to carcinoma (non-progressing dysplasia). Two pathologists independently analyzed the presence of tumor-infiltrating lymphocytes in LDy pre-embedded paraffin-fixed specimens. The RNA-based next-generation sequencing panel OIRRA was used to evaluate the expression of 395 genes related to immune system activation. (3) Results: High TILs are significantly correlated with a higher risk of malignant transformation. The non-brisk pattern was significantly associated with an 86% reduced risk of malignant progression (OR = 0.16, 95% CI: 0.03-0.5, p = 0.008). TILs showed a highly positive correlation with CCR6, CD83, HLA-DPB1, MX1 and SNAI1, and they were inversely correlated with CD48, CIITA, CXCR4, FCER1G, IL1B, LST1 and TLR8. (4) Conclusions: TILs have a great potential to identify high-risk progression dysplasia and thus to define surveillance protocols and prevention programs.
Collapse
Affiliation(s)
- Francesco Chu
- Division of Otolaryngology and Head and Neck Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Fausto Maffini
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Daniela Lepanto
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Davide Vacirca
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Sergio Vincenzo Taormina
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Rita De Berardinis
- Division of Otolaryngology and Head and Neck Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Correspondence: (R.D.B.); (M.T.); Tel.: +39-02-57489380 (R.D.B. & M.T.)
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Silvano Vignati
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Alberto Ranghiero
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Alessandra Rappa
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Susanna Chiocca
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Massimo Barberis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Marta Tagliabue
- Division of Otolaryngology and Head and Neck Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Correspondence: (R.D.B.); (M.T.); Tel.: +39-02-57489380 (R.D.B. & M.T.)
| | - Mohssen Ansarin
- Division of Otolaryngology and Head and Neck Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
15
|
Abedi-Gaballu F, Kamal Kazemi E, Salehzadeh SA, Mansoori B, Eslami F, Emami A, Dehghan G, Baradaran B, Mansoori B, Cho WC. Metabolic Pathways in Breast Cancer Reprograming: An Insight to Non-Coding RNAs. Cells 2022; 11:cells11192973. [PMID: 36230935 PMCID: PMC9563138 DOI: 10.3390/cells11192973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/10/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer cells reprogram their metabolisms to achieve high energetic requirements and produce precursors that facilitate uncontrolled cell proliferation. Metabolic reprograming involves not only the dysregulation in glucose-metabolizing regulatory enzymes, but also the enzymes engaging in the lipid and amino acid metabolisms. Nevertheless, the underlying regulatory mechanisms of reprograming are not fully understood. Non-coding RNAs (ncRNAs) as functional RNA molecules cannot translate into proteins, but they do play a regulatory role in gene expression. Moreover, ncRNAs have been demonstrated to be implicated in the metabolic modulations in breast cancer (BC) by regulating the metabolic-related enzymes. Here, we will focus on the regulatory involvement of ncRNAs (microRNA, circular RNA and long ncRNA) in BC metabolism, including glucose, lipid and glutamine metabolism. Investigation of this aspect may not only alter the approaches of BC diagnosis and prognosis, but may also open a new avenue in using ncRNA-based therapeutics for BC treatment by targeting different metabolic pathways.
Collapse
Affiliation(s)
- Fereydoon Abedi-Gaballu
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | - Elham Kamal Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | - Seyed Ahmad Salehzadeh
- Department of Medicinal Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 175-14115, Iran
| | - Behnaz Mansoori
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 175-14115, Iran
| | - Farhad Eslami
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | - Ali Emami
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51666-16471, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran
| | - Behzad Mansoori
- Cellular and Molecular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
- Correspondence: (B.M.); (W.C.C.)
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
- Correspondence: (B.M.); (W.C.C.)
| |
Collapse
|
16
|
Ding Y, Ge Y, Wang D, Liu Q, Sun S, Hua L, Deng J, Luan S, Cheng H, Xie Q, Gong Y, Zhang T. LncRNA-IMAT1 Promotes Invasion of Meningiomas by Suppressing KLF4/hsa-miR22-3p/Snai1 Pathway. Mol Cells 2022; 45:388-402. [PMID: 35680373 PMCID: PMC9200663 DOI: 10.14348/molcells.2022.2232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/18/2021] [Accepted: 12/26/2021] [Indexed: 11/27/2022] Open
Abstract
Malignant meningiomas often show invasive growth that makes complete tumor resection challenging, and they are more prone to recur after radical resection. Invasive meningioma associated transcript 1 (IMAT1) is a long noncoding RNA located on Homo sapiens chromosome 17 that was identified by our team based on absolute expression differences in invasive and non-invasive meningiomas. Our studies indicated that IMAT1 was highly expressed in invasive meningiomas compared with non-invasive meningiomas. In vitro studies showed that IMAT1 promoted meningioma cell invasion through the inactivation of the Krüppel-like factor 4 (KLF4)/hsa-miR22-3p/Snai1 pathway by acting as a sponge for hsa-miR22-3p, and IMAT1 knockdown effectively restored the tumor suppressive properties of KLF4 by preserving its tumor suppressor pathway. In vivo experiments confirmed that IMAT1 silencing could significantly inhibit the growth of subcutaneous tumors and prolong the survival period of tumor-bearing mice. Our findings demonstrated that the high expression of IMAT1 is the inherent reason for the loss of the tumor suppressive properties of KLF4 during meningioma progression. Therefore, we believe that IMAT1 may be a potential biological marker and treatment target for meningiomas.
Collapse
Affiliation(s)
- Yaodong Ding
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yu Ge
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Daijun Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qin Liu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shuchen Sun
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lingyang Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jiaojiao Deng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shihai Luan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Haixia Cheng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qing Xie
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ye Gong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Tao Zhang
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
17
|
Popov A, Mandys V. Senescence-Associated miRNAs and Their Role in Pancreatic Cancer. Pathol Oncol Res 2022; 28:1610156. [PMID: 35570840 PMCID: PMC9098800 DOI: 10.3389/pore.2022.1610156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/12/2022] [Indexed: 01/17/2023]
Abstract
Replicative senescence is irreversible cell proliferation arrest for somatic cells which can be circumvented in cancers. Cellular senescence is a process, which may play two opposite roles. On the one hand, this is a natural protection of somatic cells against unlimited proliferation and malignant transformation. On the other hand, cellular secretion caused by senescence can stimulate inflammation and proliferation of adjacent cells that may promote malignancy. The main genes controlling the senescence pathways are also well known as tumor suppressors. Almost 140 genes regulate both cellular senescence and cancer pathways. About two thirds of these genes (64%) are regulated by microRNAs. Senescence-associated miRNAs can stimulate cancer progression or act as tumor suppressors. Here we review the role playing by senescence-associated miRNAs in development, diagnostics and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Alexey Popov
- Department of Pathology, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czechia
| | | |
Collapse
|
18
|
Zhang L, Zhang Z, Xie J, Zhao Y, Tian G, Jiang H, Tao H, Liu J. Target invasion-triggered signal amplification based on duplex-specific nuclease for selective and sensitive detection of miRNAs. Anal Chim Acta 2022; 1189:339182. [PMID: 34815041 DOI: 10.1016/j.aca.2021.339182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 01/10/2023]
Abstract
Dysregulation of MicroRNAs (miRNAs) cause various diseases in humans, and developing reliable methods to detect miRNAs is critical for molecular diagnostics and personalized medicine. This study developed a toehold-mediated target invasion combined with duplex-specificity nuclease (DSN)-assisted cyclic signal amplification fluorescent sensor. Herein, we take advantage of toehold-mediated target invasion process to ensure the high selectivity of miRNA determination, coupled with the unique cleavage properties of DSN to improve the sensitivity of the strategy significantly. Throughout the assay, the whole procedure of detection the target let-7a has a limit of detection (LOD) as low as 9.00 fM and an excellent linear range from 1 pM to 100 nM for no more than 60 min. The assay shows reasonable specificity in detecting mismatched miRNAs and can realize single-base discrimination in the let-7 families. Finally, the developed method was applied to detect the miRNAs extracted from human serum. The results were consistent with those based on the quantitative reverse transcription-polymerase chain reaction(qRT-PCR) method, which shows great potential application value in clinical molecular diagnostics and biological research.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China
| | - Zhang Zhang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China
| | - Jingling Xie
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China
| | - Yuanqing Zhao
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China
| | - Hui Jiang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China
| | - Hualin Tao
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China.
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical, University, Luzhou, 646000, PR China.
| |
Collapse
|
19
|
Gao M, Mei C, Guo Y, Xia P, Zhang H, Liu Y, Yao Y, Jiang X, Yuan Y, Qian Y. Loss of FBP1 by aPKC-ι/Snail Pathway-Mediated Repression Promotes Invasion and Aerobic Glycolysis of Intrahepatic Cholangiocarcinoma. Front Oncol 2021. [DOI: 10.3389/fonc.2021.756419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is one of the most commonly diagnosed malignancies worldwide, and the second most common primary liver tumor. The lack of effective diagnostic and treatment methods results in poor patient prognosis and high mortality rate. Atypical protein kinase C-ι (aPKC-ι) is highly expressed in primary and metastatic ICC tissues, and regulates epithelial mesenchymal transition (EMT) through the aPKC-ι/P-Sp1/Snail signaling pathway. Recent studies have correlated aberrant glucose metabolism with EMT. Given the vital role of FBP1 in regulating glucose metabolism in cancer cells, we hypothesized that aPKC-ι downregulates FBP1 in ICC cells through the Snai1 pathway, and enhances glycolysis and metastasis. We confirmed the ability of aPKC-ι promotes glycolysis, invasion and metastasis of cancer cells, and further demonstrated that FBP1 inhibits the malignant properties of ICC cells by antagonizing aPKC-ι. Our findings provide novel insights into the molecular mechanisms of ICC progression and metastasis, as well as a theoretical basis for exploring new treatment strategies.
Collapse
|
20
|
Otmani K, Lewalle P. Tumor Suppressor miRNA in Cancer Cells and the Tumor Microenvironment: Mechanism of Deregulation and Clinical Implications. Front Oncol 2021; 11:708765. [PMID: 34722255 PMCID: PMC8554338 DOI: 10.3389/fonc.2021.708765] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/27/2021] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that have been identified as important posttranscriptional regulators of gene expression. miRNAs production is controlled at multiple levels, including transcriptional and posttranscriptional regulation. Extensive profiling studies have shown that the regulation of mature miRNAs expression plays a causal role in cancer development and progression. miRNAs have been identified to act as tumor suppressors (TS) or as oncogenes based on their modulating effect on the expression of their target genes. Upregulation of oncogenic miRNAs blocks TS genes and leads to tumor formation. In contrast, downregulation of miRNAs with TS function increases the translation of oncogenes. Several miRNAs exhibiting TS properties have been studied. In this review we focus on recent studies on the role of TS miRNAs in cancer cells and the tumor microenvironment (TME). Furthermore, we discuss how TS miRNA impacts the aggressiveness of cancer cells, with focus of the mechanism that regulate its expression. The study of the mechanisms of miRNA regulation in cancer cells and the TME may paved the way to understand its critical role in the development and progression of cancer and is likely to have important clinical implications in a near future. Finally, the potential roles of miRNAs as specific biomarkers for the diagnosis and the prognosis of cancer and the replacement of tumor suppressive miRNAs using miRNA mimics could be promising approaches for cancer therapy.
Collapse
Affiliation(s)
- Khalid Otmani
- Experimental Hematology Laboratory, Jules Bordet Institute, Université libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
21
|
Chirshev E, Suzuki T, Wang H, Nguyen A, Hojo N, Sanderman L, Mirshahidi S, Ioffe YJ, Unternaehrer JJ. Let-7i Reduces Aggressive Phenotype and Induces BRCAness in Ovarian Cancer Cells. Cancers (Basel) 2021; 13:cancers13184617. [PMID: 34572843 PMCID: PMC8468164 DOI: 10.3390/cancers13184617] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022] Open
Abstract
High-grade serous carcinoma of the ovary is a deadly gynecological cancer with poor long-term survival. Dysregulation of microRNAs has been shown to contribute to the formation of cancer stem cells (CSCs), an important part of oncogenesis and tumor progression. The let-7 family of microRNAs has previously been shown to regulate stemness and has tumor suppressive actions in a variety of cancers, including ovarian. Here, we demonstrate tumor suppressor actions of let-7i: repression of cancer cell stemness, inhibition of migration and invasion, and promotion of apoptosis, features important for cancer progression, relapse, and metastasis. Let-7i over-expression results in increased sensitivity to the PARP inhibitor olaparib in samples without BRCA mutations, consistent with induction of BRCAness phenotype. We also show that let-7i inhibits the expression of several factors involved in the homologous recombination repair (HRR) pathway, providing potential mechanisms by which the BRCAness phenotype could be induced. These actions of let-7i add to the rationale for use of this miRNA as a treatment for ovarian cancer patients, including those without mutations in the HRR pathway.
Collapse
Affiliation(s)
- Evgeny Chirshev
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Tise Suzuki
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Hanmin Wang
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Anthony Nguyen
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Nozomi Hojo
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Linda Sanderman
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
- Biology Department, California State University San Bernardino, San Bernardino, CA 92407, USA
| | - Saied Mirshahidi
- Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Basic Sciences, Division of Microbiology & Molecular Genetics, Loma Linda University, Loma Linda, CA 92354, USA;
| | - Yevgeniya J. Ioffe
- Department of Gynecology and Obstetrics, Division of Gynecologic Oncology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA;
| | - Juli J. Unternaehrer
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
- Department of Gynecology and Obstetrics, Loma Linda University, Loma Linda, CA 92354, USA
- Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Correspondence:
| |
Collapse
|
22
|
De Santis C, Götte M. The Role of microRNA Let-7d in Female Malignancies and Diseases of the Female Reproductive Tract. Int J Mol Sci 2021; 22:ijms22147359. [PMID: 34298978 PMCID: PMC8305730 DOI: 10.3390/ijms22147359] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
microRNAs are small noncoding RNAs that regulate gene expression at the posttranscriptional level. Let-7d is a microRNA of the conserved let-7 family that is dysregulated in female malignancies including breast cancer, ovarian cancer, endometrial cancer, and cervical cancer. Moreover, a dysregulation is observed in endometriosis and pregnancy-associated diseases such as preeclampsia and fetal growth restriction. Let-7d expression is regulated by cytokines and steroids, involving transcriptional regulation by OCT4, MYC and p53, as well as posttranscriptional regulation via LIN28 and ADAR. By downregulating a wide range of relevant mRNA targets, let-7d affects cellular processes that drive disease progression such as cell proliferation, apoptosis (resistance), angiogenesis and immune cell function. In an oncological context, let-7d has a tumor-suppressive function, although some of its functions are context-dependent. Notably, its expression is associated with improved therapeutic responses to chemotherapy in breast and ovarian cancer. Studies in mouse models have furthermore revealed important roles in uterine development and function, with implications for obstetric diseases. Apart from a possible utility as a diagnostic blood-based biomarker, pharmacological modulation of let-7d emerges as a promising therapeutic concept in a variety of female disease conditions.
Collapse
MESH Headings
- Aging
- Animals
- Biomarkers
- Biomarkers, Tumor
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Cell Line, Tumor
- Female
- Fertility/genetics
- Gene Expression Regulation
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Genital Diseases, Female/drug therapy
- Genital Diseases, Female/genetics
- Genital Neoplasms, Female/drug therapy
- Genital Neoplasms, Female/genetics
- Humans
- Mice
- MicroRNAs/genetics
- MicroRNAs/physiology
- Molecular Targeted Therapy
- Pregnancy
- Pregnancy Complications/genetics
- RNA, Neoplasm/antagonists & inhibitors
- RNA, Neoplasm/genetics
- RNA, Neoplasm/physiology
Collapse
|