1
|
Zhao Y, Shi G, Huang X, Zhang Z, Liao K, Xiong H, Feng Z, Mao S, Zhang X. LRP8 inhibits bladder cancer cell ferroptosis by activating the Wnt/β-catenin-SCD1 positive feedback loop. Hum Mol Genet 2025; 34:843-851. [PMID: 39989004 DOI: 10.1093/hmg/ddaf024] [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: 04/22/2024] [Revised: 01/17/2025] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Advanced bladder cancer (bc) patients often have poor prognoses due to issues such as recurrence and drug resistance. The discovery of ferroptosis has opened new avenues for bc treatment; however, the specific regulatory mechanisms remain to be explored. This study aimed to investigate the mechanisms influencing ferroptosis in bc cells, with a particular focus on the role of low-density lipoprotein receptor-related protein 8 (LRP8). METHODS We utilized reverse transcription-quantitative polymerase chain reaction and western blot to assess the expression of LRP8 in bc cells, activation of the Wnt/β-catenin signaling pathway, and the expression of genes related to fatty acid synthesis. We measured changes in ferroptosis levels by evaluating mitochondrial membrane potential, Fe2+, malondialdehyde, and reactive oxygen species levels. A xenograft mouse model was employed to validate the impact of LRP8 on bc progression. RESULTS Cell experiments demonstrated a significant upregulation of LRP8 expression in bc cells. Knockdown of LRP8 induced ferroptosis in bc cells, a process directly triggered by the inhibition of the Wnt/β-catenin signaling pathway. Activation of the Wnt/β-catenin signaling pathway mediated by LRP8 upregulated the expression of stearoyl-CoA desaturase 1 (SCD1), subsequently leading to the suppression of ferroptosis. In vivo experiments indicated that LRP8 knockdown significantly impaired bc growth, accompanied by inhibition of the Wnt/β-catenin-SCD1 axis. CONCLUSION LRP8 mediates the synthesis of monounsaturated fatty acids through the Wnt/β-catenin-SCD1 positive feedback loop, thereby inhibiting ferroptosis in bc cells. These findings provide a promising target for the regulation of ferroptosis in bc cells.
Collapse
Affiliation(s)
- Yong Zhao
- Department of Urology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Guohong Shi
- Department of Urology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Xiang Huang
- Department of Urology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Zhongyuan Zhang
- Department of Pathology, Nanping First Hospital of Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Kaijun Liao
- Department of Pharmacy, Nanping First Hospital of Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Hao Xiong
- Department of Urology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Zhiqiang Feng
- Department of Urology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Shihui Mao
- Department of Urology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| | - Xu Zhang
- Department of Urology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317 Zhongshan Road, Yanping District, Nanping City, Fujian Province 353000, China
| |
Collapse
|
2
|
Wang R, Wang F. CircCENPM serves as a CeRNA to aggravate nasopharyngeal carcinoma metastasis and stemness via enhancing BMI1. Hereditas 2025; 162:39. [PMID: 40087716 PMCID: PMC11907939 DOI: 10.1186/s41065-025-00406-7] [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: 09/29/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a malignant head and neck cancer with high mortality and dismal prognosis. Emerging research have disclosed that circRNAs are crucial gene expression regulators engaged in tumor advancement. This work aspired to identify novel oncogenic circRNA driving NPC progression. METHODS Bioinformatics analysis was performed to explore and predict underlying circRNA and downstream targets. Luciferase reporter assay was executed to check the binding relationship between these genes. Cell function tests were conducted using CCK-8, would healing, and flow cytometry. The stemness markers CD133, Nanog and Oct4 was detected via western blot. RESULTS CircCENPM was notably enhanced in NPC. Silencing of circCENPM suppressed NPC cell growth, migration, and stemness in vitro, simultaneously impeded tumorigenesis of NPC in vivo. Moreover, circCENPM could interact with miR-362-3p, whereas miR-362-3p inhibitor apparently reversed the mitigated growth and stemness induced by circCENPM knockdown in NPC cells. Furthermore, BMI1 was identified to be the downstream target of miR-362-3p, and BMI1 introduction partially offset the anti-tumor function of miR-362-3p in NPC cells. CONCLUSION CircCENPM functioned as a carcinogenic driver and facilitated NPC growth and stemness via miR-362-3p/BMI1 regulatory network, which provided a potential biomarker and attractive target for NPC intervention and treatment.
Collapse
Affiliation(s)
- Rui Wang
- The Second Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Fei Wang
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China.
| |
Collapse
|
3
|
Liu B, Bukhari I, Li F, Ren F, Xia X, Hu B, Liu H, Meyer TF, Marshall BJ, Tay A, Fu Y, Wu W, Tang Y, Mi Y, Zheng PY. Enhanced LRP8 expression induced by Helicobacter pylori drives gastric cancer progression by facilitating β-Catenin nuclear translocation. J Adv Res 2025; 69:299-312. [PMID: 38609049 PMCID: PMC11954824 DOI: 10.1016/j.jare.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
INTRODUCTION Helicobacter pylori (H. pylori) infection has been associated with gastric carcinogenesis. However, the precise involvement of LRP8, the low-density lipoprotein receptor-related protein 8, in H. pylori pathogenesis and gastric cancer (GC) remains poorly understood. OBJECTIVES To investigate the potential role of LRP8 in H. pylori infection and gastric carcinogenesis. METHODS Three-dimensional human-derived gastric organoids (hGO) and gastric cancer organoids (hGCO) were synthesized from the tissues obtained from human donors. In this work, multi-omics combined with in vivo and in vitro studies were conducted to investigate the potential involvement of LRP8 in H. pylori-induced GC. RESULTS We found that H. pylori infection significantly upregulated the expression of LRP8 in human GC tissues, cells, organoids, and mouse gastric mucous. In particular, LRP8 exhibited a distinct enrichment in cancer stem cells (CSC). Functionally, silencing of LRP8 affected the formation and proliferation of tumor spheroids, while increased expression of LRP8 was associated with increased proliferation and stemness of GC cells and organoids. Mechanistically, LRP8 promotes the binding of E-cadherin to β-catenin, thereby promoting nuclear translocation and transcriptional activity of β-catenin. Furthermore, LRP8 interacts with the cytotoxin-associated gene A (CagA) to form the CagA/LRP8/β-catenin complex. This complex further amplifies H. pylori-induced β-catenin nuclear translocation, leading to increased transcription of inflammatory factors and CSC markers. Clinical analysis demonstrated that abnormal overexpression of LRP8 is correlated with a poor prognosis and resistance to 5-Fluorouracil in patients with GC. CONCLUSION Our findings provide valuable information on the molecular intricacies of H. pylori-induced gastric carcinogenesis, offering potential therapeutic targets and prognostic markers for GC.
Collapse
Affiliation(s)
- Bin Liu
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Ihtisham Bukhari
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Fazhan Li
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Feifei Ren
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xue Xia
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Baitong Hu
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Haipeng Liu
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Thomas F Meyer
- Max Planck Institute for Infection Biology, Department of Molecular Biology, 10117 Berlin, Germany; Laboratory of Infection Oncology, Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts University of Kiel, Kiel, Germany
| | - Barry J Marshall
- Helicobacter Pylori Research Laboratory, School of Biomedical Sciences, Marshall Centre for Infectious Disease Research and Training, University of Western Australia, Nedlands 6009, Australia
| | - Alfred Tay
- Helicobacter Pylori Research Laboratory, School of Biomedical Sciences, Marshall Centre for Infectious Disease Research and Training, University of Western Australia, Nedlands 6009, Australia
| | - Yuming Fu
- Gastrointestinal Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wanqing Wu
- Gastrointestinal Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Youcai Tang
- Department of Pediatrics, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Peng-Yuan Zheng
- Henan Key Laboratory for Helicobacter pylori and Digestive Tract Microecology, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
4
|
Sandhanam K, Tamilanban T, Bhattacharjee B, Manasa K. Exploring miRNA therapies and gut microbiome-enhanced CAR-T cells: advancing frontiers in glioblastoma stem cell targeting. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2169-2207. [PMID: 39382681 DOI: 10.1007/s00210-024-03479-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024]
Abstract
Glioblastoma multiforme (GBM) presents a formidable challenge in oncology due to its aggressive nature and resistance to conventional treatments. Recent advancements propose a novel therapeutic strategy combining microRNA-based therapies, chimeric antigen receptor-T (CAR-T) cells, and gut microbiome modulation to target GBM stem cells and transform cancer treatment. MicroRNA therapies show promise in regulating key signalling pathways implicated in GBM progression, offering the potential to disrupt GBM stem cell renewal. CAR-T cell therapy, initially successful in blood cancers, is being adapted to target GBM by genetically engineering T cells to recognise and eliminate GBM stem cell-specific antigens. Despite early successes, challenges like the immunosuppressive tumour microenvironment persist. Additionally, recent research has uncovered a link between the gut microbiome and GBM, suggesting that gut dysbiosis can influence systemic inflammation and immune responses. Novel strategies to modulate the gut microbiome are emerging, enhancing the efficacy of microRNA therapies and CAR-T cell treatments. This combined approach highlights the synergistic potential of these innovative therapies in GBM treatment, aiming to eradicate primary tumours and prevent recurrence, thereby improving patient prognosis and quality of life. Ongoing research and clinical trials are crucial to fully exploit this promising frontier in GBM therapy, offering hope to patients grappling with this devastating disease.
Collapse
Affiliation(s)
- K Sandhanam
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, 603203, Tamil Nadu, India
| | - T Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, 603203, Tamil Nadu, India.
| | - Bedanta Bhattacharjee
- Department of Pharmacology, Girijananda Chowdhury University-Tezpur Campus, 784501, Assam, India
| | - K Manasa
- Department of Pharmacology, MNR College of Pharmacy, Sangareddy, 502294, Telangana, India
| |
Collapse
|
5
|
Xu Y, Zhou Y, Yi X, Nie X. LRP8 promotes tumorigenesis in ovarian cancer through inhibiting p53 signaling. Cell Biol Int 2024; 48:626-637. [PMID: 38263609 DOI: 10.1002/cbin.12133] [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: 09/06/2023] [Revised: 12/13/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024]
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy with a high mortality rate. Low-density lipoprotein (LDL) receptor-related protein 8 (LRP8) is a cell membrane receptor belonging LDL receptor family and is involved in several tumor progressions. However, there is limited understanding of how LRP8 mediates OC development. LRP8 expression level was identified in human OC tissues and cells using immunohistochemical staining and quantitative polymerase chain reaction assays, respectively. Functions of LRP8 in OC progression were evaluated by Celigo cell counting, wound healing, transwell and flow cytometry assays, and the xenograft models. The human phospho-kinase array analysis was used for screening potential signaling involved in OC development. We observed that LRP8 was overexpressed in OC tissues, and high expression of LRP8 was associated with poor prognosis of OC patients. Functionally, LRP8 knockdown remarkably reduced proliferation and migration of OC cells, and induced apoptosis and S phase cycle arrest. LRP8 deficiency attenuated in vivo tumor growth of OC cells. Moreover, the addition of p53 inhibitor partially reversed the effects of LRP8 knockdown on OC cell proliferation and apoptosis, indicating the involvement of p53 signaling in LRP8-mediated OC progression. This study confirmed that LRP8/p53 axis contributed to OC progression, which might serve as a novel potential therapeutic target for OC patients.
Collapse
Affiliation(s)
- Yan Xu
- Department of Obstetrics and Gynecology, Shenyang Women's and Children's Hospital, Shenyang, China
| | - Yang Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiling Yi
- Department of Obstetrics and Gynecology, Shenyang Women's and Children's Hospital, Shenyang, China
| | - Xiaocui Nie
- Department of Obstetrics and Gynecology, Shenyang Women's and Children's Hospital, Shenyang, China
| |
Collapse
|
6
|
Hu Y, Li J, Liu C, Zhang X, Wang Y, Lin J, Peng Z, Zhu L. MiR362-3p Alleviates Osteosarcoma by Regulating the IL6ST/JAK2/STAT3 Pathway in Vivo and in Vitro. Technol Cancer Res Treat 2024; 23:15330338241261616. [PMID: 39051528 PMCID: PMC11273602 DOI: 10.1177/15330338241261616] [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: 01/21/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 07/27/2024] Open
Abstract
Objectives: To investigate the effects and the related signaling pathway of miR-362-3p on OS. Methods: The bioinformatics analysis approaches were employed to investigate the target pathway of miR-362-3p. After the 143B and U2OS cells and nu/nu male mice were randomly divided into blank control (BC) group, normal control (NC) group, and overexpression group (OG), the CCK-8, EdU staining, wound healing assay, Transwell assay, and TUNEL staining were adopted to respectively determine the effects of overexpressed miR-362-3p on the cell viability, proliferation, migration, invasion, and apoptosis of 143B and U2OS cells in vitro, tumor area assay and hematoxylin and eosin staining were employed to respectively determine the effects of overexpressed miR-362-3p on the growth and pathological injury of OS tissue in vivo. The qRT-PCR, Western blot, and immunohistochemical staining were applied to respectively investigate the effects of overexpressed miR-362-3p on the IL6ST/JAK2/STAT3 pathway in OS in vivo and in vitro. Results: The bioinformatics analysis approaches combined qRT-PCR indicated that the IL6ST/JAK2/STAT3 is one of the target pathways of miR-362-3p. Compared with NC, the cell viability, proliferation, migration, and invasion of 143B and U2OS cells were dramatically (P < 0.01) inhibited but the apoptosis was prominently (P <0 .0001) promoted in OG. Compared with NC, the growth of OS tissue was significantly (P < 0.05) suppressed and the pathological injury of OS tissue was substantially aggravated in OG. The gene expression levels of IL6ST, JAK2, and STAT3 and the protein expression levels of IL6ST, JAK2, p-JAK2, STAT3, and p-STAT3 in 143B and U2OS cells were memorably (P < 0.0001) lower in OG than those in NC. In addition, the positively stained areas of proteins of IL6ST, JAK2, p-JAK2, STAT3, and p-STAT3 of OS tissue in OG were markedly (P < 0.01) reduced compared with those in NC. Conclusion: The overexpression of miR362-3p alleviates OS by inhibiting the IL6ST/JAK2/STAT3 pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Yunteng Hu
- Department of Spine Surgery, Zhujiang Hosptial, Southern Medical University, Guangzhou, China
| | - Jianjun Li
- Department of Spine Surgery, Zhujiang Hosptial, Southern Medical University, Guangzhou, China
| | - Chun Liu
- Department of Spine Surgery, Zhujiang Hosptial, Southern Medical University, Guangzhou, China
| | - Xue Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Yihan Wang
- Department of Spine Surgery, Zhujiang Hosptial, Southern Medical University, Guangzhou, China
| | - Jiezhao Lin
- Department of Spine Surgery, Zhujiang Hosptial, Southern Medical University, Guangzhou, China
| | - Ziyue Peng
- Department of Spine Surgery, Zhujiang Hosptial, Southern Medical University, Guangzhou, China
| | - Lixin Zhu
- Department of Spine Surgery, Zhujiang Hosptial, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Zhang J, Shang L, Jiang W, Wu W. Shikonin induces apoptosis and autophagy via downregulation of pyrroline-5-carboxylate reductase1 in hepatocellular carcinoma cells. Bioengineered 2022; 13:7904-7918. [PMID: 35293266 PMCID: PMC9208523 DOI: 10.1080/21655979.2022.2052673] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Shikonin(SK) is a natural small molecule naphthoquinone compound, which has anti-cancer activity in various human malignant tumors. Pyrroline-5-carboxylate reductase 1(PYCR1) is involved in tumorigenesis and regulates various cellular processes, including growth, invasion, migration, and apoptosis. However, the effect of SK and PYCR1 on apoptosis and autophagy in hepatocellular carcinoma are unclear. Our goal is to determine the internal molecular mechanism of the interaction between SK and PYCR1 and its role in the occurrence and development of liver cancer. The CCK8 assay, wound healing assay, and transwell assays show that SK and siPYCR1(gene silence PYCR1) inhibited the malignant phenotype of HCC cells, including cell viability, colony formation, migration, and invasion, respectively. The flow cytometry assays and immunofluorescence show that SK and siPYCR1 activated apoptosis and autophagy, respectively. SK induces apoptosis and autophagy in a dose-dependent manner. In addition, HCC cells were transfected with small interference fragment PYCR1 siRNA to construct siPYCR1 and SK single treatment group and co-treatment group to verify the interaction between SK and PYCR1. The Western blot identified that PI3K/Akt/mTOR signal pathway protein expression was significantly downregulated in HCC cells treated with SK and siPYCR1 together. Collectively, SK may induce apoptosis and autophagy by reducing the expression of PYCR1 and suppressing PI3K/Akt/mTOR. Thus, SK may be a promising antineoplastic drug in Hepatocellular carcinoma (HCC). SK downregulating PYCR1 might supply a theoretical foundation for the potential therapeutic application in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Junli Zhang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Ling Shang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Wendi Jiang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Wenjuan Wu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| |
Collapse
|
8
|
Rizk NI, Abulsoud AI, Kamal MM, Kassem DH, Hamdy NM. Exosomal-long non-coding RNAs journey in colorectal cancer: Evil and goodness faces of key players. Life Sci 2022; 292:120325. [PMID: 35031258 DOI: 10.1016/j.lfs.2022.120325] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 02/07/2023]
Abstract
Exosomes are nano-vesicles (NVs) secreted by cells and take part in cell-cell communications. Lately, these exosomes were proved to have dual faces in cancer. Actually, they can contribute to carcinogenesis through epithelial-mesenchymal transition (EMT), angiogenesis, metastasis and tumor microenvironment (TME) of various cancers, including colorectal cancer (CRC). On the other hand, they can be potential targets for cancer treatment. CRC is one of the most frequent tumors worldwide, with incidence rates rising in the recent decades. In its early stage, CRC is asymptomatic with poor treatment outcomes. Therefore, finding a non-invasive, early diagnostic biomarker tool and/or suitable defender to combat CRC is mandatory. Exosomes provide enrichment and safe setting for their cargos non-coding RNAs (ncRNAs) and proteins, whose expression levels can be upregulated ordown-regulated in cancer. Hence, exosomes can be used as diagnostic and/or prognostic tools for cancer. Moreover, exosomes can provide a novel potential therapeutic modality for tumors via loading with specific chemotherapeutic agents, with the advantage of possible tumor targeting. In this review, we will try to collect and address recent studies concerned with exosomes and their cargos' implications for CRC diagnosis and/or hopefully, treatment.
Collapse
Affiliation(s)
- Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy (Boys Branch), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Mohamed M Kamal
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt; The Centre for Drug Research and Development, Faculty of Pharmacy, BUE, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|