1
|
Hsu CY, Altalbawy FMA, Oghenemaro EF, Uthirapathy S, Chandra M, Nathiya D, Kaur P, Ravi Kumar M, Kadhim AJ, Kariem M. Exosomal lncRNAs in the Tumor Angiogenesis: As Therapeutic Targets in Cancer Treatment. Arch Pharm (Weinheim) 2025; 358:e202400940. [PMID: 40165644 DOI: 10.1002/ardp.202400940] [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: 12/12/2024] [Revised: 02/12/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Exosomes, as mediators of intercellular communication, can be released from different types of cells and regulate the function of the target cell by transferring cargo, such as proteins, DNA, and RNA. Recent investigations have revealed a preponderance of long noncoding RNAs (lncRNAs), a subclass of noncoding RNAs, within exosomes, where they exhibit notable stability and are implicated in the development and progression of neoplastic processes, such as tumor angiogenesis. Angiogenesis, as a hallmark of cancer, provides diffusible nutrients and oxygen to the distant cells and guarantees tumorigenesis and metastasis. Exosomal lncRNAs, including MALAT1, OIP5-AS1, PART1, SNHG family, FAM225A, ATB, RAMP2-AS1, UCA1, TRPM2-AS, FGD5-AS1, and LINC0016, could modulate tumor angiogenesis by activating signaling cascades and mediators within the target cells, such as microRNAs (miRNAs). Regulation of tumor angiogenesis through modulation of exosomal lncRNAs could be a reliable strategy for cancer therapy. In this review, we discuss the characteristics and biogenesis of exosomes and lncRNAs and how exosomal lncRNAs are involved in various processes of tumorigenesis. Our primary focus is on exosomal lncRNAs, their impact on tumor angiogenesis, and their potential as novel diagnostic markers and therapeutic targets for various cancers.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, Egypt
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
2
|
Zhang T, Lu C, Lv M, Du S, Wu X. NOL6 Promotes Tumor Progression by Facilitating Cancer Cell-Induced Platelet Aggregation and Angiogenesis in Breast Cancer. FRONT BIOSCI-LANDMRK 2025; 30:25361. [PMID: 40152371 DOI: 10.31083/fbl25361] [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: 06/24/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND Breast cancer (BC) is a prevalent malignancy among women, and numerous investigations have reported that platelet aggregation may play a role in BC progression. Thus, identifying new targets for BC is essential. In this regard, we focused on nucleolar protein 6 (NOL6), located on chromosome 9p13, which is implicated in tumor development. OBJECTIVE To investigate NOL6 expression in BC, examine its role in platelet aggregation and angiogenesis, and elucidate the underlying mechanisms. METHODS Bioinformatic analyses, immunoblotting, and quantitative real-time polymerase chain reaction (qPCR) were performed to assess NOL6 expression in BC. Cell counting kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assays were conducted to determine the impact of NOL6 on BC cell proliferation. Immunostaining, enzyme-linked immunosorbent assay (ELISA), and flow cytometry (FCM) assays were utilized to analyze the effects of NOL6 on platelet aggregation. Tube formation and transwell assays were performed to examine angiogenesis and invasion, immunoblot assays were used to confirm the underlying mechanisms, and tumor growth assays in mice were conducted to validate the findings in vivo. RESULTS NOL6 was found to be highly expressed in BC and was associated with patient prognosis, platelet aggregation, and angiogenesis. Its knockdown inhibited BC cell proliferation and reduced platelet aggregation induced by BC cells. Additionally, NOL6 depletion impaired angiogenesis and migration of BC cells. In vivo studies confirmed that NOL6 promotes tumor growth. Mechanistically, NOL6 enhances the Twisted spiral transcription factor 1 (Twist1)/galectin-3 axis, contributing to BC progression. CONCLUSIONS NOL6 can promote tumor progression by facilitating platelet aggregation and angiogenesis in BC cells through the Twist1/galectin-3 axis.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of General Surgery, Lianyungang Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, 222000 Lianyungang, Jiangsu, China
| | - Cheng Lu
- Department of Breast, Nanjing Maternal and Child Health Hospital, 210094 Nanjing, Jiangsu, China
| | - Mingming Lv
- Department of Breast, Nanjing Maternal and Child Health Hospital, 210094 Nanjing, Jiangsu, China
| | - Shengwang Du
- Department of General Surgery, Lianyungang Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, 222000 Lianyungang, Jiangsu, China
| | - Xinjun Wu
- Department of General Surgery, Lianyungang Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, 222000 Lianyungang, Jiangsu, China
| |
Collapse
|
3
|
El-Ashmawy NE, Khedr EG, Abo-Saif MA, Hamouda SM. Cuproptosis regulation by long noncoding RNAs: Mechanistic insights and clinical implications in cancer. Arch Biochem Biophys 2025; 765:110324. [PMID: 39900259 DOI: 10.1016/j.abb.2025.110324] [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: 12/13/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Although survival rates have been improved in recent years, the prognosis of many cancer types remains inadequate, mostly owing to treatment resistance. Moreover, there is a continued need for exploring novel and reliable tumor markers to achieve accurate diagnosis. Understanding the molecular complexity of cancer allows for the development of more effective and personalized treatments and facilitates the discovery of biomarkers that surpass traditional ones and assist in cancer diagnosis and monitoring disease progression and response to treatment. Recent studies exploring the complexity of cancer biology have identified a new form of cell death, known as cuproptosis, which is driven by the accumulation of copper and subsequent stress induced by dysregulation of copper homeostasis. Increased copper level enables cancer cells to maintain their accelerated growth rates and metastatic potential, yet these cells can evade cuproptosis. Long noncoding RNAs (lncRNAs) have been recognized for their pivotal role in different hallmarks of cancer, including resistance to cell death. They have been found to be implicated in controlling copper balance and cuproptosis. Besides, lncRNAs associated with cuproptosis pathway have demonstrated their potential as diagnostic and prognostic cancer biomarkers as well as indicators of treatment response. Our review aims to summarize recent studies focusing on the intricate relationship between lncRNAs and cuproptosis and explore the mechanisms by which lncRNAs can modulate copper homeostasis and regulate cuproptosis pathway. We also highlight recent discoveries concerning the role of cuproptosis-related lncRNAs in diagnosis, prognosis, and therapy of different types of cancer. By elucidating the significance of cuproptosis-related lncRNAs, this review provides insights into how these lncRNAs can be used to develop new therapeutic strategies to improve treatment outcomes.
Collapse
Affiliation(s)
- Nahla E El-Ashmawy
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk, Cairo, Postal Code: 11837, Egypt.
| | - Eman G Khedr
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt.
| | - Mariam A Abo-Saif
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt.
| | - Sara M Hamouda
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, El-Gharbia, Tanta, Postal Code: 31527, Egypt.
| |
Collapse
|
4
|
Shi M, Zhang R, Lyu H, Xiao S, Guo D, Zhang Q, Chen XZ, Tang J, Zhou C. Long non-coding RNAs: Emerging regulators of invasion and metastasis in pancreatic cancer. J Adv Res 2025:S2090-1232(25)00073-6. [PMID: 39933650 DOI: 10.1016/j.jare.2025.02.001] [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: 11/09/2024] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND The invasion and metastasis of pancreatic cancer (PC) are key factors contributing to disease progression and poor prognosis. This process is primarily driven by EMT, which has been the focus of recent studies highlighting the role of long non-coding RNAs (lncRNAs) as crucial regulators of EMT. However, the mechanisms by which lncRNAs influence invasive metastasis are multifaceted, extending beyond EMT regulation alone. AIM OF REVIEW This review primarily aims to characterize lncRNAs affecting invasion and metastasis in pancreatic cancer. We summarize the regulatory roles of lncRNAs across multiple molecular pathways and highlight their translational potential, considering the implications for clinical applications in diagnostics and therapeutics. KEY SCIENTIFIC CONCEPTS OF REVIEW The review focuses on three principal scientific themes. First, we primarily summarize lncRNAs orchestrate various signaling pathways, such as TGF-β/Smad, Wnt/β-catenin, and Notch, to regulate molecular changes associated with EMT, thereby enhancing cellular motility and invasivenes. Second, we summarize the effects of lncRNAs on autophagy and ferroptosis and discuss the role of exosomal lncRNAs in the tumor microenvironment to regulate the behavior of neighboring cells and promote cancer cell invasion. Third, we emphasize the effects of RNA modifications (such as m6A and m5C methylation) on stabilizing lncRNAs and enhancing their capacity to mediate invasive metastasis in PC. Lastly, we discuss the translational potential of these findings, emphasizing the inherent challenges in using lncRNAs as clinical biomarkers and therapeutic targets, while proposing prospective research strategies.
Collapse
Affiliation(s)
- Mengmeng Shi
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qi Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China.
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China.
| |
Collapse
|
5
|
Mai X, Chen X, Wang Z, Xian H, Wen Q, Sun G, Wang T. Screening Differentially Expressed Proteins in Areca Nut-Related Oral Squamous Cell Carcinoma Using Tandem Mass Tag Proteomics. Int Dent J 2025:S0020-6539(24)01635-6. [PMID: 39757032 DOI: 10.1016/j.identj.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE Areca nut chewing has been associated with a poor prognosis in oral squamous cell carcinoma (OSCC). This study seeks to identify differentially expressed proteins among areca nut-related OSCC,non-areca nut-related OSCC and adjacent normal epithelial tissues, with the aim of providing novel insights for the investigation of areca nut-related OSCC. METHODS Using Tandem mass tag (TMT)-based proteomic analysis, a comparative proteomic profiling was conducted among areca nut-related OSCC, non-areca nut-related OSCC, and adjacent normal epithelial tissues (n=15). GO and KEGG enrichment analyses were then employed to identify significant proteins pertinent to the pathogenesis of OSCC for further study. Western Blot (WB) and Immunohistochemistry (IHC) techniques were used to preliminary validate the expression patterns of LZTS1, MMP10, MYH6, MB, and TNNC1 among various groups (n=30). RESULTS 27 differentially expressed proteins were identified when comparing the areca nut-related OSCC group with both the non-areca nut-related OSCC and normal epithelial tissue groups. Among these, 15 proteins were upregulated, while 12 were downregulated. LZTS1 and MMP10 were included in the upregulated proteins, whereas MYH6, MB, and TNNC1 were downregulated. WB and IHC analyses corroborated the proteomic findings, revealing consistent expression trends for these 5 proteins across the studied groups. CONCLUSION LZTS1, MMP10, MYH6, MB and TNNC1 emerge as promising biomarkers for assessing disease progression, prognosis, and potential targeted therapies in areca nut-related OSCC.
Collapse
Affiliation(s)
- Xiaoqun Mai
- Department of stomatology, Hainan General Hospital, Haikou, China; The Affiliated Hainan Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xinyu Chen
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China.
| | - Zihan Wang
- School of Stomatology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haiyu Xian
- Department of stomatology, Hainan General Hospital, Haikou, China; The Affiliated Hainan Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Qitao Wen
- Department of stomatology, Hainan General Hospital, Haikou, China; The Affiliated Hainan Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Guanyu Sun
- Department of stomatology, Hainan General Hospital, Haikou, China; The Affiliated Hainan Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Tao Wang
- Department of stomatology, Hainan General Hospital, Haikou, China; The Affiliated Hainan Hospital of Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
6
|
Zhang JX, Shen YB, Ma DD, Li ZH, Zhang ZY, Jin WD. LINC01857 promotes cell proliferation and migration while dampening cell apoptosis in pancreatic cancer by upregulating CDC42EP3 via miR-450b-5p. Heliyon 2024; 10:e38427. [PMID: 39524859 PMCID: PMC11547965 DOI: 10.1016/j.heliyon.2024.e38427] [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: 05/28/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024] Open
Abstract
Background Pancreatic cancer (PC) is a devastating human malignancy with a poor survival outcome (5-year survival less than 10 %). In recent years, the regulatory roles of long non-coding RNAs (lncRNAs) in various types of cancers have been widely reported. Based on bioinformatics analysis, LINC01857 is shown to be highly expressed in PC tissue. Nevertheless, the role of LINC01857 in PC is limitedly reported. Hence, this study aimed to explore the effects of lncRNA LINC01857 on PC cell process and the related mechanism. Methods RT-qPCR and fluorescence in situ hybridization (FISH) assay were conducted to measure LINC01857 expression and distribution in PANC-1 and MIA PaCa-2 cells. Colony formation and wound healing assays as well as flow cytometry analyses were employed to estimate the proliferation, migration, and apoptosis of PC cells transfected with pcDNA3.1-LINC01857 or si-LINC01857 compared with the behavior of PC cells transfected with empty pcDNA3.1 vector (control) or si-negative control (NC). Furthermore, RNA pulldown and luciferase reporter assays were utilized to demonstrate the interaction of LINC01857 and miR-450b-5p or to validate the binding of miR-450b-5p and cell division cycle 42 effector protein 3 (CDC42EP3). Results LINC01857 was highly expressed in PANC-1 and MIA PaCa-2 cells in contrast to its expression in pancreatic ductal epithelial cells (8.9 folds and 7.1 folds, p < 0.001). Silencing LINC01857 significantly reduced cell proliferation and migration while enhancing apoptosis (p < 0.0005). In contrast, overexpression of LINC01857 markedly (p < 0.05) accelerated these malignant behavior of PC cells. MiR-450b-5p was targeted and inversely regulated by LINC01857. Moreover, CDC42EP3 was verified to be targeted by miR-450b-5p, and CDC42EP3 was correlated to LINC01857 in a positive manner (p < 0.001). Rescue experiments manifested that silencing CDC42EP3 effectively (p < 0.05) reversed the promoting effect of LINC01857 on malignant behavior of PC cells. Conclusion LINC01857 promotes PC cell proliferation and migration while obstructing cell apoptosis by binding to miR-450b-5p and thus regulating CDC42EP3 expression. The study presents a novel and promising regulatory axis, which holds potential for the identification of biomarkers and development of therapeutic strategies for PC treatment.
Collapse
Affiliation(s)
| | | | - Dan-Dan Ma
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| | - Zhong-Hu Li
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| | - Zhi-Yong Zhang
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| | - Wei-Dong Jin
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| |
Collapse
|
7
|
Bustamante A, Baritaki S, Zaravinos A, Bonavida B. Relationship of Signaling Pathways between RKIP Expression and the Inhibition of EMT-Inducing Transcription Factors SNAIL1/2, TWIST1/2 and ZEB1/2. Cancers (Basel) 2024; 16:3180. [PMID: 39335152 PMCID: PMC11430682 DOI: 10.3390/cancers16183180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Untreated primary carcinomas often lead to progression, invasion and metastasis, a process that involves the epithelial-to-mesenchymal transition (EMT). Several transcription factors (TFs) mediate the development of EMT, including SNAIL1/SNAIL2, TWIST1/TWIST2 and ZEB1/ZEB2, which are overexpressed in various carcinomas along with the under expression of the metastasis suppressor Raf Kinase Inhibitor Protein (RKIP). Overexpression of RKIP inhibits EMT and the above associated TFs. We, therefore, hypothesized that there are inhibitory cross-talk signaling pathways between RKIP and these TFs. Accordingly, we analyzed the various properties and biomarkers associated with the epithelial and mesenchymal tissues and the various molecular signaling pathways that trigger the EMT phenotype such as the TGF-β, the RTK and the Wnt pathways. We also presented the various functions and the transcriptional, post-transcriptional and epigenetic regulations for the expression of each of the EMT TFs. Likewise, we describe the transcriptional, post-transcriptional and epigenetic regulations of RKIP expression. Various signaling pathways mediated by RKIP, including the Raf/MEK/ERK pathway, inhibit the TFs associated with EMT and the stabilization of epithelial E-Cadherin expression. The inverse relationship between RKIP and the TF expressions and the cross-talks were further analyzed by bioinformatic analysis. High mRNA levels of RKIP correlated negatively with those of SNAIL1, SNAIL2, TWIST1, TWIST2, ZEB1, and ZEB2 in several but not all carcinomas. However, in these carcinomas, high levels of RKIP were associated with good prognosis, whereas high levels of the above transcription factors were associated with poor prognosis. Based on the inverse relationship between RKIP and EMT TFs, it is postulated that the expression level of RKIP in various carcinomas is clinically relevant as both a prognostic and diagnostic biomarker. In addition, targeting RKIP induction by agonists, gene therapy and immunotherapy will result not only in the inhibition of EMT and metastases in carcinomas, but also in the inhibition of tumor growth and reversal of resistance to various therapeutic strategies. However, such targeting strategies must be better investigated as a result of tumor heterogeneities and inherent resistance and should be better adapted as personalized medicine.
Collapse
Affiliation(s)
- Andrew Bustamante
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Apostolos Zaravinos
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
Gao Y, Shen Y, Dong J, Zhou Y, Zhu C, Yu Q, Qin X. Pancreatic head carcinoma derived from the dorsal pancreas is more likely to metastasize early than from the ventral pancreas through microvascular invasion. Medicine (Baltimore) 2024; 103:e39296. [PMID: 39151507 PMCID: PMC11332757 DOI: 10.1097/md.0000000000039296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/15/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
The development of the pancreatic head originates from the fusion of the ventral and dorsal pancreatic primordia during embryonic development. Theoretically, the origin of pancreatic head cancer also exists from the ventral pancreas and the dorsal pancreas. Among 49 patients with pancreatic head cancer, pancreatic head cancer was divided into pancreatic head cancer originating from the ventral (PHCv) or dorsal pancreas (PHCd) through imaging and pathological classification. The clinical data was collected and compared between the PHCv group and the PHCd group. The results showed that the patients from the PHCd group had worse long-term survival than those from the PHCv group (10 months vs 14.5 months). Similarly, the progression-free survival (PFS) results also indicate that patients from the PHCd group had a shorter time than those from the PHCv group (5 months vs 9.5 months). Further stratified analysis of potentially related factors showed that microvascular invasion is related to poor prognosis, and patients with pancreatic head cancer derived from the dorsal pancreas are more likely to develop microvascular invasion.
Collapse
Affiliation(s)
- Yuan Gao
- The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
- Department of Hepato-biliary-pancreatic Surgery, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| | - Yuhang Shen
- The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| | - Jun Dong
- The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| | - Yang Zhou
- Department of Pathology, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| | - Chunfu Zhu
- Department of Hepato-biliary-pancreatic Surgery, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| | - Qiang Yu
- Department of Hepato-biliary-pancreatic Surgery, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| | - Xihu Qin
- The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
- Department of Hepato-biliary-pancreatic Surgery, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| |
Collapse
|
9
|
Cong L, Shi J, Zhao J, Li K, Dai D, Zhang B, Zhao W. Huaier inhibits cholangiocarcinoma cells through the twist1/FBP1/Wnt/β-catenin axis. Mol Biol Rep 2024; 51:842. [PMID: 39042261 DOI: 10.1007/s11033-024-09738-5] [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: 02/02/2024] [Accepted: 06/18/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Although Huaier granules can be used as prospective anti-cholangiocarcinoma drugs, the mechanism of action of Huaier granules in cholangiocarcinoma is not clear. The anti-cholangiocarcinoma effect of Huaier granules was validated in cell line research. In vitro experiments were conducted to investigate the signalling pathways affected by Huaier in CCA cells. METHODS AND RESULTS Real-time quantitative PCR (RT‒qPCR) and Western blot analysis were performed to analyse gene expression in CCA cells. MTT assays, scratch tests, and Transwell assays were used to explore the effects on the proliferation and metastasis of CCA cells. Chromatin immunoprecipitation assays were performed to reveal the potential underlying mechanisms involved. Twist1 was upregulated in human CCA tissues. In addition, its expression levels were negatively related to FBP1 expression levels. Mechanistically, Twist1 can bind to the region of the FBP1 promoter to reduce its expression. Huaier plays an indispensable role in suppressing Twist1 expression to inhibit the Twist1/FBP1/Wnt/β-catenin axis. Then, we verified the effect of Huaier in vitro. CONCLUSIONS These findings suggested that Huaier granules were capable of inhibiting CCA development through regulating the Twist1/FBP1/Wnt/β-catenin signalling axis and provided a novel orientation for the development of novel anti-CCA drugs.
Collapse
Affiliation(s)
- Liyuan Cong
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266003, People's Republic of China
| | - Jian Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266003, People's Republic of China
| | - Jing Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, People's Republic of China
| | - Kun Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266003, People's Republic of China
| | - Dongdong Dai
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266003, People's Republic of China
| | - Bingyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266003, People's Republic of China
| | - Wei Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266003, People's Republic of China.
| |
Collapse
|
10
|
Liu ZY, Tang JM, Yang MQ, Yang ZH, Xia JZ. The role of LncRNA-mediated autophagy in cancer progression. Front Cell Dev Biol 2024; 12:1348894. [PMID: 38933333 PMCID: PMC11199412 DOI: 10.3389/fcell.2024.1348894] [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: 12/03/2023] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) are a sort of transcripts that are more than 200 nucleotides in length. In recent years, many studies have revealed the modulatory role of lncRNAs in cancer. Typically, lncRNAs are linked to a variety of essential events, such as apoptosis, cellular proliferation, and the invasion of malignant cells. Simultaneously, autophagy, an essential intracellular degradation mechanism in eukaryotic cells, is activated to respond to multiple stressful circumstances, for example, nutrient scarcity, accumulation of abnormal proteins, and organelle damage. Autophagy plays both suppressive and promoting roles in cancer. Increasingly, studies have unveiled how dysregulated lncRNAs expression can disrupt autophagic balance, thereby contributing to cancer progression. Consequently, exploring the interplay between lncRNAs and autophagy holds promising implications for clinical research. In this manuscript, we methodically compiled the advances in the molecular mechanisms of lncRNAs and autophagy and briefly summarized the implications of the lncRNA-mediated autophagy axis.
Collapse
Affiliation(s)
- Zi-yuan Liu
- Gastroenterological Surgery, The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University, Wuxi, China
- Department of General Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Jia-ming Tang
- Department of Neurology, The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Meng-qi Yang
- Gastroenterological Surgery, The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University, Wuxi, China
- Department of General Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Zhi-hui Yang
- Department of General Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Jia-zeng Xia
- Gastroenterological Surgery, The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University, Wuxi, China
- Department of General Surgery, Jiangnan University Medical Center, Wuxi, China
| |
Collapse
|
11
|
Huang Y, Zhang R, Lyu H, Xiao S, Guo D, Chen XZ, Zhou C, Tang J. LncRNAs as nodes for the cross-talk between autophagy and Wnt signaling in pancreatic cancer drug resistance. Int J Biol Sci 2024; 20:2698-2726. [PMID: 38725864 PMCID: PMC11077374 DOI: 10.7150/ijbs.91832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/06/2024] [Indexed: 05/12/2024] Open
Abstract
Pancreatic cancer is a malignancy with high mortality. In addition to the few symptoms until the disease reaches an advanced stage, the high fatality rate is attributed to its rapid development, drug resistance and lack of appropriate treatment. In the selection and research of therapeutic drugs, gemcitabine is the first-line drug for pancreatic cancer. Solving the problem of gemcitabine resistance in pancreatic cancer will contribute to the progress of pancreatic cancer treatment. Long non coding RNAs (lncRNAs), which are RNA transcripts longer than 200 nucleotides, play vital roles in cellular physiological metabolic activities. Currently, our group and others have found that some lncRNAs are aberrantly expressed in pancreatic cancer cells, which can regulate the process of cancer through autophagy and Wnt/β-catenin pathways simultaneously and affect the sensitivity of cancer cells to therapeutic drugs. This review presents an overview of the recent evidence concerning the node of lncRNA for the cross-talk between autophagy and Wnt/β-catenin signaling in pancreatic cancer, together with the practicability of lncRNAs and the core regulatory factors as targets in therapeutic resistance.
Collapse
Affiliation(s)
- Yuhan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada, T6G2R3
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China, 430068
| |
Collapse
|
12
|
Liu K, Chen H, Li Y, Wang B, Li Q, Zhang L, Liu X, Wang C, Ertas YN, Shi H. Autophagy flux in bladder cancer: Cell death crosstalk, drug and nanotherapeutics. Cancer Lett 2024; 591:216867. [PMID: 38593919 DOI: 10.1016/j.canlet.2024.216867] [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: 01/21/2024] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Autophagy, a self-digestion mechanism, has emerged as a promising target in the realm of cancer therapy, particularly in bladder cancer (BCa), a urological malignancy characterized by dysregulated biological processes contributing to its progression. This highly conserved catabolic mechanism exhibits aberrant activation in pathological events, prominently featured in human cancers. The nuanced role of autophagy in cancer has been unveiled as a double-edged sword, capable of functioning as both a pro-survival and pro-death mechanism in a context-dependent manner. In BCa, dysregulation of autophagy intertwines with cell death mechanisms, wherein pro-survival autophagy impedes apoptosis and ferroptosis, while pro-death autophagy diminishes tumor cell survival. The impact of autophagy on BCa progression is multifaceted, influencing metastasis rates and engaging with the epithelial-mesenchymal transition (EMT) mechanism. Pharmacological modulation of autophagy emerges as a viable strategy to impede BCa progression and augment cell death. Notably, the introduction of nanoparticles for targeted autophagy regulation holds promise as an innovative approach in BCa suppression. This review underscores the intricate interplay of autophagy with cell death pathways and its therapeutic implications in the nuanced landscape of bladder cancer.
Collapse
Affiliation(s)
- Kuan Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Huijing Chen
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Yanhong Li
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Bei Wang
- Department of Gynecology, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Qian Li
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Lu Zhang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Xiaohui Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China.
| | - Ce Wang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, 38039, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Turkey; UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, 06800, Turkey.
| | - Hongyun Shi
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China.
| |
Collapse
|
13
|
Mo Y, Adu-Amankwaah J, Qin W, Gao T, Hou X, Fan M, Liao X, Jia L, Zhao J, Yuan J, Tan R. Unlocking the predictive potential of long non-coding RNAs: a machine learning approach for precise cancer patient prognosis. Ann Med 2023; 55:2279748. [PMID: 37983519 PMCID: PMC11571739 DOI: 10.1080/07853890.2023.2279748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023] Open
Abstract
The intricate web of cancer biology is governed by the active participation of long non-coding RNAs (lncRNAs), playing crucial roles in cancer cells' proliferation, migration, and drug resistance. Pioneering research driven by machine learning algorithms has unveiled the profound ability of specific combinations of lncRNAs to predict the prognosis of cancer patients. These findings highlight the transformative potential of lncRNAs as powerful therapeutic targets and prognostic markers. In this comprehensive review, we meticulously examined the landscape of lncRNAs in predicting the prognosis of the top five cancers and other malignancies, aiming to provide a compelling reference for future research endeavours. Leveraging the power of machine learning techniques, we explored the predictive capabilities of diverse lncRNA combinations, revealing their unprecedented potential to accurately determine patient outcomes.
Collapse
Affiliation(s)
- Yixuan Mo
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, China
| | - Wenjie Qin
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, China
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
| | - Tan Gao
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
| | - Xiaoqing Hou
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
| | - Mengying Fan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
| | - Xuemei Liao
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
| | - Liwei Jia
- Department of Pathology, UT Southwestern Medical Center, Dallas, UT, USA
| | - Jinming Zhao
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Pathology, The First Hospital of China Medical University, Shenyang, China
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
| | - Rubin Tan
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
14
|
Hu J, Liu J, Zhou S, Luo H. A review on the role of gamma-butyrobetaine hydroxylase 1 antisense RNA 1 in the carcinogenesis and tumor progression. Cancer Cell Int 2023; 23:263. [PMID: 37925403 PMCID: PMC10625699 DOI: 10.1186/s12935-023-03113-3] [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: 08/02/2023] [Accepted: 10/27/2023] [Indexed: 11/06/2023] Open
Abstract
Gamma-butyrobetaine hydroxylase 1 antisense RNA 1 (BBOX1-AS1), located on human chromosome 11 p14, emerges as a critical player in tumorigenesis with diverse oncogenic effects. Aberrant expression of BBOX1-AS1 intricately regulates various cellular processes, including cell growth, epithelial-mesenchymal transition, migration, invasion, metastasis, cell death, and stemness. Notably, the expression of BBOX1-AS1 was significantly correlated with clinical-pathological characteristics and tumor prognoses, and it could also be used for the diagnosis of lung and esophageal cancers. Through its involvement in the ceRNA network, BBOX1-AS1 competitively binds to eight miRNAs in ten different cancer types. Additionally, BBOX1-AS1 can directly modulate downstream protein-coding genes or act as an mRNA stabilizer. The implications of BBOX1-AS1 extend to critical signaling pathways, including Hedgehog, Wnt/β-catenin, and MELK/FAK pathways. Moreover, it influences drug resistance in hepatocellular carcinoma. The present study provides a systematic review of the clinical significance of BBOX1-AS1's aberrant expression in diverse tumor types. It sheds light on the intricate molecular mechanisms through which BBOX1-AS1 influences cancer initiation and progression and outlines potential avenues for future research in this field.
Collapse
Affiliation(s)
- Juan Hu
- Medical Service Division, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Jipeng Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330000, Jiangxi, People's Republic of China
| | - Siwei Zhou
- Second School of Clinical Medicine, Nanchang University, Nanchang, 330038, Jiangxi, China
| | - Hongliang Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330000, Jiangxi, People's Republic of China.
| |
Collapse
|
15
|
Mu J, Jin H, Wu H. Effects of nutritional therapy on gastrointestinal microbial digestion and barrier defense markers in elderly patients with diabetes. Aging Clin Exp Res 2023; 35:2667-2674. [PMID: 37821689 DOI: 10.1007/s40520-023-02518-4] [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: 05/30/2023] [Accepted: 07/25/2023] [Indexed: 10/13/2023]
Abstract
OBJECTIVE We sought to investigate the effects of gastrointestinal nutrition therapy on gastrointestinal microbial digestion and barrier defense markers in elderly patients with diabetes. METHODS A total of 120 elderly patients with type 2 diabetes were enrolled at our hospital between January 2020 and December 2022. The participants in this study were randomly allocated into either the nutritional group (n = 60) who underwent gastrointestinal nutrition therapy or the control group (n = 60) who underwent conventional T2DM diet management for a period of 12 weeks. Clinical data, as well as small intestinal permeability measured by the lactulose-mannitol urine test, plasma circulating IL-6 and zonulin levels measured by ELISA, and expressions of ZO-1 and Claudin-3 in blood analyzed through Western blotting were collected. RESULTS The nutrition group demonstrated a higher proportion of patients achieving HbA1c < 7% compared to the control group (P < 0.05). Moreover, the nutrition group exhibited a greater reduction in fasting and postprandial blood glucose levels compared to the control group (P < 0.05). The concentrations of formate-tetrahydrofolate ligase and acetic CoA transferase were significantly increased in the nutrition group compared to the control group (P < 0.05). Fecal analysis revealed higher levels of acetic acid and butyric acid in the nutrition group compared to the control group (P < 0.05). The ratio of lactulose to mannitol was higher in the nutrition group compared to the control group (P < 0.05). Furthermore, the nutrition group showed lower levels of IL-6 and zonulin compared to the control group (P < 0.05). CONCLUSION Personalized gastrointestinal nutrition therapy was found to enhance the production of short-chain fatty acids and preserve intestinal permeability, leading to improved gastrointestinal microbial digestion and barrier defense in elderly patients with diabetes.
Collapse
Affiliation(s)
- Jianfo Mu
- Data Analysis Center, Shanghai Healink Medical Information Consulting Co., LTD, Beijing, China
| | - Hefeng Jin
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Hui Wu
- Research Center, Shanghai Healing Medical Information Consulting Co., LTD, No.258 Pingyang Road, Minhang District, Shanghai, 201102, China.
| |
Collapse
|
16
|
Kyrgiafini MA, Giannoulis T, Chatziparasidou A, Christoforidis N, Mamuris Z. Unveiling the Genetic Complexity of Teratozoospermia: Integrated Genomic Analysis Reveals Novel Insights into lncRNAs' Role in Male Infertility. Int J Mol Sci 2023; 24:15002. [PMID: 37834450 PMCID: PMC10573971 DOI: 10.3390/ijms241915002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Male infertility is a global health issue, affecting over 20 million men worldwide. Genetic factors are crucial in various male infertility forms, including teratozoospermia. Nonetheless, the genetic causes of male infertility remain largely unexplored. In this study, we employed whole-genome sequencing and RNA expression analysis to detect differentially expressed (DE) long-noncoding RNAs (lncRNAs) in teratozoospermia, along with mutations that are exclusive to teratozoospermic individuals within these DE lncRNAs regions. Bioinformatic tools were used to assess variants' impact on lncRNA structure, function, and lncRNA-miRNA interactions. Our analysis identified 1166 unique mutations in teratozoospermic men within DE lncRNAs, distinguishing them from normozoospermic men. Among these, 64 variants in 23 lncRNAs showed potential regulatory roles, 7 variants affected 4 lncRNA structures, while 37 variants in 17 lncRNAs caused miRNA target loss or gain. Pathway Enrichment and Gene Ontology analyses of the genes targeted by the affected miRNAs revealed dysregulated pathways in teratozoospermia and a link between male infertility and cancer. This study lists novel variants and lncRNAs associated for the first time with teratozoospermia. These findings pave the way for future studies aiming to enhance diagnosis and therapy in the field of male infertility.
Collapse
Affiliation(s)
- Maria-Anna Kyrgiafini
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| | - Themistoklis Giannoulis
- Laboratory of Biology, Genetics and Bioinformatics, Department of Animal Sciences, University of Thessaly, Gaiopolis, 41336 Larissa, Greece
| | - Alexia Chatziparasidou
- Embryolab IVF Unit, St. 173-175 Ethnikis Antistaseos, Kalamaria, 55134 Thessaloniki, Greece
| | | | - Zissis Mamuris
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| |
Collapse
|