1
|
Duran T, Balikci I, Buyukkosucu B, Gunes IF, Pekgonul HK, Vardar N, Yilmaz MD, Ak G, Zengin G. Biological Characterization of One Oxadiazole Derivative (5(4-Hydroxyphenyl)-2-(N-Phenyl Amino)-1,3,4-Oxadiazole): In Vitro, In Silico, and Network Pharmacological Approaches. Chem Biol Drug Des 2025; 105:e70038. [PMID: 39757393 DOI: 10.1111/cbdd.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Oxadiazole compounds are of great interest because they have a range of biological activities ranging from antioxidants to anticancer agents. Against this background, we wanted to demonstrate the antioxidant, enzyme inhibitory, and anticancer effects of 5(4-hydroxyphenyl)-2-(N-phenylamino)-1,3,4-oxadiazole (Hppo). Antioxidant abilities were measured through free radical scavenging and reducing power tests. Enzyme inhibitory effects were studied by cholinesterases, tyrosinase, amylase, and glucosidase. The anticancer effect was tested on pancreatic cancer cell lines (PANC-1, CRL-169) and on HEK293 cell lines. The compound showed significant antioxidant activity (particularly in the CUPRAC (cupric acid-reducing antioxidant capacity) assay) and enzyme inhibitory properties (particularly glucosidase inhibition). In the anticancer test, the compound showed strong anticancer activity in pancreatic cancer with apoptotic signaling pathways. These results were confirmed by molecular modeling and bioinformatics tools. Thus, our findings can provide novel and versatile compounds for the development of multidirectional drugs in the pharmaceutical industry.
Collapse
Affiliation(s)
- Tugce Duran
- Department of Medical Genetics, Faculty of Medicine, KTO Karatay University, Konya, Turkey
- Department of Pediatric Allergy and Immunology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Irem Balikci
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Busra Buyukkosucu
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Ibrahim Furkan Gunes
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Hatice Kubra Pekgonul
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Necati Vardar
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
- Department of Metallurgy and Materials Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Mahmut Deniz Yilmaz
- Department of Basic Sciences, Faculty of Engineering, Necmettin Erbakan University, Konya, Turkey
- BITAM-Science and Technology Research and Application Center, Necmettin Erbakan University, Konya, Turkey
| | - Gunes Ak
- Department of Biology, Faculty of Science, Selcuk University, Konya, Turkey
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University, Konya, Turkey
| |
Collapse
|
2
|
Anand S, Patel TN. Integrating the metabolic and molecular circuits in diabetes, obesity and cancer: a comprehensive review. Discov Oncol 2024; 15:779. [PMID: 39692821 DOI: 10.1007/s12672-024-01662-1] [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: 10/04/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024] Open
Abstract
The progressive globalization of sedentary lifestyles and diets rich in lipids and processed foods has caused two major public health hazards-diabetes and obesity. The strong interlink between obesity and type 2 diabetes mellitus and their combined burden encompass them into a single term 'Diabesity'. They have also been tagged as the drivers for the onset of cancer. The clinical association between diabetes, obesity, and several types of human cancer demands an assessment of vital junctions correlating the three. This review focuses on revisiting the molecular axis linking diabetes and obesity to cancer through pathways that get imbalanced owing to metabolic upheaval. We also attempt to describe the functional disruptions of DNA repair mechanisms due to overwhelming oxidative DNA damage caused by diabesity. Genomic instability, a known cancer hallmark results when DNA repair does not work optimally, and as will be inferred from this review the obtruded metabolic homeostasis in diabetes and obesity creates a favorable microenvironment supporting metabolic reprogramming and enabling malignancies. Altered molecular and hormonal landscapes in these two morbidities provide a novel connection between metabolomics and oncogenesis. Understanding various aspects of the tumorigenic process in diabesity-induced cancers might help in the discovery of new biomarkers and prompt targeted therapeutic interventions.
Collapse
Affiliation(s)
- Shrikirti Anand
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Trupti N Patel
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
3
|
Hashemi M, Rezaei M, Rezaeiaghdam H, Jamali B, Koohpar ZK, Tanha M, Bizhanpour A, Asadi S, Jafari AM, Khosroshahi EM, Eslami M, Salimimoghadam S, Nabavi N, Rashidi M, Fattah E, Taheriazam A, Entezari M. Highlighting function of Wnt signalling in urological cancers: Molecular interactions, therapeutic strategies, and (nano)strategies. Transl Oncol 2024; 50:102145. [PMID: 39357465 PMCID: PMC11474201 DOI: 10.1016/j.tranon.2024.102145] [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: 10/25/2023] [Revised: 05/06/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
Cancer is a complex, multistep process characterized by abnormal cell growth and metastasis as well as the capacity of the tumor cells in therapy resistance development. The urological system is particularly susceptible to a group of malignancies known as urological cancers, where an accumulation of genetic alterations drives carcinogenesis. In various human cancers, Wnt singalling is dysregulated; following nuclear transfer of β-catenin, it promotes tumor progression and affects genes expression. Elevated levels of Wnt have been documented in urological cancers, where its overexpression enhances growth and metastasis. Additionally, increased Wnt singalling contributes to chemoresistance in urological cancers, leading to reduced sensitivity to chemotherapy agents like cisplatin, doxorubicin, and paclitaxel. Wnt upregulation can change radiotherapy response of urological cancers. The regulation of Wnt involves various molecular pathways, including Akt, miRNAs, lncRNAs, and circRNAs, all of which play roles in carcinogenesis. Targeting and silencing Wnt or its associated pathways can mitigate tumorigenesis in urological cancers. Anti-cancer compounds such as curcumin and thymoquinone have shown efficacy in suppressing tumorigenesis through the downregulation of Wnt singalling. Notably, nanoparticles have proven effective in treating urological cancers, with several studies in prostate cancer (PCa) using nanoparticles to downregulate Wnt and suppress tumor growth. Future research should focus on developing small molecules that inhibit Wnt singalling to further suppress tumorigenesis and advance the treatment of urological cancers. Moreover, Wnt can be used as reliable biomarker for the diagnosis and prognosis of urological cancers.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Rezaei
- Health Research Center, Chamran Hospital, Tehran, Iran
| | - Hadi Rezaeiaghdam
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Behdokht Jamali
- Department of Microbiology and Genetics, Kherad Institute of Higher Education, Bushehr, Iran
| | - Zeinab Khazaei Koohpar
- Department Of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Mahsa Tanha
- Department Of Biological Sciences, University Of Alabama, Tuscaloosa, Al, United States
| | - Anahita Bizhanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Maedeh Eslami
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8V 1P7, Canada
| | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Eisa Fattah
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Xiang Z, Wang Y, Ma X, Song S, He Y, Zhou J, Feng L, Yang S, Wu Y, Yu B, Xia G, Xu W, Zhao Y, Wang L. Targeting the NOTCH2/ADAM10/TCF7L2 Axis-Mediated Transcriptional Regulation of Wnt Pathway Suppresses Tumor Growth and Enhances Chemosensitivity in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2405758. [PMID: 39601111 DOI: 10.1002/advs.202405758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/12/2024] [Indexed: 11/29/2024]
Abstract
Wnt/β-catenin/transcription factor (TCF) transcriptional activity plays an integral role in colorectal cancer (CRC) carcinogenesis. However, to date, no drugs targeting this pathway are used in clinical practice owing to the undesirable and serious side effects. In this study, it is found that the transcriptional regulation of Wnt pathway is activated and associated with liver metastasis in CRC. Through high-throughput screening of 24 inhibitors on 12 CRC and three colorectal organoids in this organoid living biobank, adavivint is found to exhibit anti-tumor activity and low toxicity in colorectal organoids, independent of the canonical Wnt/β-catenin signaling. Mechanistically, ADAM10 is screened as a target of adavivint to specifically regulate the protein expression of NOTCH2, which mediates the transcriptional regulation of the Wnt pathway. NOTCH2 not directly interact with TCF7-like 2 (TCF7L2), a key downstream transcriptional factor of canonical Wnt/β-catenin signaling, but directly activated the transcription of TCF7L2 and Wnt target genes, such as MYC, JUN and CCND1/2. Furthermore, use of adavivint or blockage of ADAM10/NOTCH2/TCF7L2 signaling enhances the chemosensitivity of CRC cells. Overall, this study provides a promising candidate for the development of small-molecule inhibitors and reveals a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhen Xiang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Yiwei Wang
- Department of general surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, P. R. China
| | - Xiao Ma
- Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, P. R. China
| | - Shuzheng Song
- Department of Colorectal Surgery, Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, P. R. China
| | - Yuanqiao He
- Center of Laboratory Animal Science, Nanchang University, No.999, Xuefu Road, Nanchang, 330031, P. R. China
| | - Jiamin Zhou
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Longhai Feng
- Department of Colorectal Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), 1 Banshan East Road, Hangzhou, 310022, P. R. China
| | - Su Yang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yibin Wu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Bingran Yu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Guangkai Xia
- Department of general surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, P. R. China
| | - Weiqi Xu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Yiming Zhao
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| | - Lu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai, 200032, China
| |
Collapse
|
5
|
Lan S, Liu S, Wang K, Chen W, Zheng D, Zhuang Y, Zhang S. tRNA-derived RNA fragment, tRF-18-8R6546D2, promotes pancreatic adenocarcinoma progression by directly targeting ASCL2. Gene 2024; 927:148739. [PMID: 38955307 DOI: 10.1016/j.gene.2024.148739] [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: 01/29/2024] [Revised: 05/11/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Pancreatic adenocarcinoma (PAAD) is a life-threatening cancer. Exploring new diagnosis and treatment targets helps improve its prognosis. tRNA-derived small non-coding RNAs (tsRNAs) are a novel type of gene expression regulators and their dysregulation is closely related to many human cancers. Yet the expression and functions of tsRNAs in PAAD are not well understood. Our study used RNA sequencing to identify tsRNA expression profiles in PAAD cells cultured in no or high glucose media and found tRF-18-8R6546D2 was an uncharacterized tsRNA, which has significantly high expression in PAAD cells and tissues. Clinically, tRF-18-8R6546D2 is linked to poor prognosis in PAAD patients and can be used to distinguish them from healthy populations. Functionally, in vitro and vivo, tRF-18-8R6546D2 over-expression promoted PAAD cell proliferation, migration and invasion, inhibited apoptosis, whereas tRF-18-8R6546D2 knock-down showed opposite effects. Mechanistically, tRF-18-8R6546D2 promoted PAAD malignancy partly by directly silencing ASCL2 and further regulating its downstream genes such as MYC and CASP3. These findings show that tRF-18-8R6546D2 is a novel oncogenic factor and can be a promising diagnostic or prognostic biomarker and therapeutic target for PAAD.
Collapse
Affiliation(s)
- Sihua Lan
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Epigenetics and Gene Regulation of Malignant Tumors, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510000, China
| | - Sixue Liu
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Ke Wang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Wenying Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Dandan Zheng
- Doctor of excellence program, First Affiliated Hospital of Jilin University, Changchun 130000, China
| | - Yanyan Zhuang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Epigenetics and Gene Regulation of Malignant Tumors, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510000, China.
| | - Shineng Zhang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Epigenetics and Gene Regulation of Malignant Tumors, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510000, China.
| |
Collapse
|
6
|
Famili-Youth EHH, Famili-Youth A, Yang D, Siddique A, Wu EY, Liu W, Resnick MB, Chen Q, Brodsky AS. Aberrant expression of collagen type X in solid tumor stroma is associated with EMT, immunosuppressive and pro-metastatic pathways, bone marrow stromal cell signatures, and poor survival prognosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.621984. [PMID: 39605631 PMCID: PMC11601388 DOI: 10.1101/2024.11.13.621984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Collagen type X (ColXα1, encoded by COL10A1) is expressed specifically in the cartilage-to-bone transition, in bone marrow cells, and in osteoarthritic (OA) cartilage. We have previously shown that ColXα1 is expressed in breast tumor stroma, correlates with tumor-infiltrating lymphocytes, and predicts poor adjuvant therapy outcomes in ER+/HER2+ breast cancer. However, the underlying molecular mechanisms for these effects are unknown. In this study, we performed bioinformatic analysis of COL10A1-associated gene modules in breast and pancreatic cancer as well as in cells from bone marrow and OA cartilage. These findings provide important insights into the mechanisms of transcriptional and extracellular matrix changes which impact the local stromal microenvironment and tumor progression. Methods Immunohistochemistry was performed to examine collagen type X expression in solid tumors. WGCNA was used to generate COL10A1-associated gene networks in breast and pancreatic tumor cohorts using RNA-Seq data from The Cancer Genome Atlas. Computational analysis was employed to assess the impact of these gene networks on development and progression of cancer and OA. Data processing and statistical analysis was performed using R and various publicly-available computational tools. Results Expression of COL10A1 and its associated gene networks highlights inflammatory and immunosuppressive microenvironments, which identify aggressive breast and pancreatic tumors and contribute to metastatic potential in a sex-dependent manner. Both cancer types are enriched in stroma, and COL10A1 implicates bone marrow-derived fibroblasts as drivers of the epithelial-to-mesenchymal transition (EMT) in these tumors. Heightened expression of COL10A1 and its associated gene networks is correlated with poorer patient outcomes in both breast and pancreatic cancer. Common transcriptional changes and chondrogenic activity are shared between cancer and OA cartilage, suggesting that similar microenvironmental alterations may underlie both diseases. Conclusions COL10A1-associated gene networks may hold substantial value as regulators and biomarkers of aggressive tumor phenotypes with implications for therapy development and clinical outcomes. Identification of tumors which exhibit high expression of COL10A1 and its associated genes may reveal the presence of bone marrow-derived stromal microenvironments with heightened EMT capacity and metastatic potential. Our analysis may enable more effective risk assessment and more precise treatment of patients with breast and pancreatic cancer.
Collapse
Affiliation(s)
- Elliot H H Famili-Youth
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Aryana Famili-Youth
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Dongfang Yang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ayesha Siddique
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Elizabeth Y Wu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Wenguang Liu
- Department of Orthopedics, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Murray B Resnick
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Qian Chen
- Department of Orthopedics, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Alexander S Brodsky
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| |
Collapse
|
7
|
Helen H, Gunawan MC, Halim P, Dinata MR, Ahmed A, Dalimunthe A, Marianne M, Ribeiro RIMDA, Hasibuan PAZ, Nurkolis F, Hey-Hawkins E, Park MN, Harahap U, Kim SH, Kim B, Syahputra RA. Flavonoids as modulators of miRNA expression in pancreatic cancer: Pathways, Mechanisms, And Therapeutic Potential. Biomed Pharmacother 2024; 179:117347. [PMID: 39241569 DOI: 10.1016/j.biopha.2024.117347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
Pancreatic cancer (PC) is a complex malignancy, distinguished by its aggressive characteristics and unfavorable prognosis. Recent developments in understanding the molecular foundations of this disease have brought attention to the noteworthy involvement of microRNAs (miRNAs) in disease development, advancement, and treatment resistance. The anticancer capabilities of flavonoids, which are a wide range of phytochemicals present in fruits and vegetables, have attracted considerable interest because of their ability to regulate miRNA expression. This review provides the effects of flavonoids on miRNA expression in PC, explains the underlying processes, and explores the possible therapeutic benefits of flavonoid-based therapies. Flavonoids inhibit PC cell proliferation, induce apoptosis, and enhance chemosensitivity via the modulation of miRNAs involved in carcinogenesis. Additionally, this review emphasizes the significance of certain miRNAs as targets of flavonoid action. These miRNAs have a role in regulating important signaling pathways such as the phosphoinositide-3-kinase-protein kinase B/Protein kinase B (Akt), mitogen activated protein kinase (MAPK), Janus kinase/signal transducers and activators of transcription (JAK/STAT), and Wnt/β-catenin pathways. This review aims to consolidate current knowledge on the interaction between flavonoids and miRNAs in PC, providing a comprehensive analysis of how flavonoid-mediated modulation of miRNA expression could influence cancer progression and therapy. It highlights the use of flavonoid nanoformulations to enhance stability, increase absorption, and maximize anti-PC activity, improving patient outcomes. The review calls for further research to optimize the use of flavonoid nanoformulations in clinical trials, leading to innovative treatment strategies and more effective approaches for PC.
Collapse
Affiliation(s)
- Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Mega Carensia Gunawan
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Muhammad Riza Dinata
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Amer Ahmed
- Department of Bioscience, Biotechnology and Environment, University of Bari, Bari, Italy
| | - Aminah Dalimunthe
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Marianne Marianne
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Rosy Iara Maciel De Azambuja Ribeiro
- Experimental Pathology Laboratory, Federal University of São João del Rei (UFSJ), 400, Sebastião Gonçalves Coelho, Chanadour, Divinópolis 35501-296, MG, Brazil
| | | | - Fahrul Nurkolis
- Biological Sciences, Faculty of Sciences and Technology, UIN Sunan Kalijaga, Yogyakarta, Indonesia
| | - Evamarie Hey-Hawkins
- Leipzig University, Faculty of Chemistry and Mineralogy, Centre for Biotechnology and Biomedicine (BBZ), Institute of Bioanalytical Chemistry, Deutscher Platz 5, 04103 Leipzig, Germany
| | - Moon Nyeo Park
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; College of Korean Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Urip Harahap
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Sung-Hoon Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Bonglee Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; College of Korean Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia.
| |
Collapse
|
8
|
Jin X, Wang S, Luo L, Yan F, He Q. Targeting the Wnt/β-catenin signal pathway for the treatment of gastrointestinal cancer: Potential for advancement. Biochem Pharmacol 2024; 227:116463. [PMID: 39102994 DOI: 10.1016/j.bcp.2024.116463] [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: 05/07/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Gastrointestinal cancers (GICs) are highly prevalent cancers that threaten human health worldwide. The Wnt/β-catenin signaling pathway has been reported to play a pivotal role in the carcinogenesis of GICs. Numerous interventions targeting the Wnt/β-catenin signaling in GICs are currently being tested in clinical trials with promising results. Unfortunately, there are no clinically approved drugs that effectively target this pathway. This comprehensive review aims to evaluate the impact of clinical therapies targeting the Wnt/β-catenin signaling pathway in GICs. By integrating data from bioinformatics databases and recent literature from the past five years, we examine the heterogeneous expression and regulatory mechanisms of Wnt/β-catenin pathway genes and proteins in GICs. Specifically, we focus on expression patterns, mutation frequencies, and clinical prognoses to understand their implications for treatment strategies. Additionally, we discuss recent clinical trial efforts targeting this pathway. Understanding the inhibitors currently under clinical investigation may help optimize foundational research and clinical strategies. We hope that elucidating the current status of precision therapeutic stratification for patients targeting the Wnt/β-catenin pathway will guide future innovations in precision medicine for GICs.
Collapse
Affiliation(s)
- Xizhi Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Fangjie Yan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China.
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
9
|
Sun R, Sun C, Yue Z, Yin G, Zhou L, Zhang S, Zhang Y, Tang D, Tan X. Astragali Radix-Curcumae Rhizoma herb pair reduces the stemness of colorectal cancer cells through HIF-2α/β-catenin pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155824. [PMID: 38941816 DOI: 10.1016/j.phymed.2024.155824] [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: 02/20/2024] [Revised: 06/02/2024] [Accepted: 06/11/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common causes of cancer-related mortality and significantly impairs quality of life. Astragali Radix-Curcumae Rhizoma (AC) is widely employed in the treatment of CRC in Chinese medicine, but the precise mechanisms remain unclear. PURPOSE This study aimed to elucidate the mechanisms by which AC inhibits CRC progression. METHODS The active components of AC were identified using UPLC-MS/MS analysis. An orthotopic transplantation colorectal tumor model was established in BALB/c mice using the CT26-Lucifer cell line to evaluate the effects of AC. Tumor volumes were monitored using IVIS imaging technology. Histological examination of tumor morphology was performed with hematoxylin and eosin (H&E) staining. Transcriptomic sequencing of mouse tumor samples was conducted to identify critical pathways and molecular targets. The impact of AC on cell viability and migration was assessed using CCK-8 and wound healing assays, respectively. To investigate the effects of AC on CRC cells, an in vitro hypoxic model was established using cobalt chloride (CoCl2), a hypoxia inducer. HIF-2α overexpression was achieved by constructing stable lentiviral vectors. Key targets identified from RNA-seq, such as c-Myc, Ki-67, β-catenin, cleaved caspase 3, CD133, and CD44, were evaluated using western blotting, qRT-PCR, and immunofluorescence assays. Epithelial-Mesenchymal Transition (EMT) and spheroid cloning assays were employed to evaluate phenotypic changes in cancer stem cells. RESULTS Twelve components of AC were identified. AC effectively inhibited CRC progression in vivo. Transcriptomic analysis highlighted hypoxic signaling as a significantly enriched pathway, implicating its role in suppressing CRC progression by AC. In the hypoxic model, AC inhibited the proliferation and migration of CRC cells in vitro. Furthermore, AC reduced cancer stemness by downregulating stemness markers, inhibiting EMT, and decreasing tumor sphere formation. The downregulation of hypoxic responses and the shift in stemness by AC involved attenuation of HIF-2α and WNT/β-catenin signaling. CONCLUSION This study provides the first evidence that AC reduces the stemness of CRC and the inhibition of the transition of CRC to stem-like cells by AC is closely related to the downregulation of the HIF-2α/β-catenin pathway, especially under hypoxic conditions.
Collapse
Affiliation(s)
- Ruiqian Sun
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Sun
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Zengyaran Yue
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Gang Yin
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lingling Zhou
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shuo Zhang
- Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Affiliated with Nanjing University of Chinese Medicine, Nantong, 226007, China
| | - Yu Zhang
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Decai Tang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiying Tan
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
10
|
Nilkhet S, Vongthip W, Lertpatipanpong P, Prasansuklab A, Tencomnao T, Chuchawankul S, Baek SJ. Ergosterol inhibits the proliferation of breast cancer cells by suppressing AKT/GSK-3beta/beta-catenin pathway. Sci Rep 2024; 14:19664. [PMID: 39179606 PMCID: PMC11344128 DOI: 10.1038/s41598-024-70516-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
Breast cancer is a prevalent malignancy affecting women globally, necessitating effective treatment strategies. This study explores the potential of ergosterol, a bioactive compound found in edible mushrooms, as a candidate for breast cancer treatment. Breast cancer cell lines (MCF-7 and MDA-MB-231) were treated with ergosterol, revealing its ability to inhibit cell viability, induce cell cycle arrest, and suppress spheroid formation. Mechanistically, ergosterol demonstrated significant inhibitory effects on the Wnt/beta-catenin signaling pathway, a critical regulator of cancer progression, by attenuating beta-catenin translocation in the nucleus. This suppression was attributed to the inhibition of AKT/GSK-3beta phosphorylation, leading to decreased beta-catenin stability and activity. Additionally, ergosterol treatment impacted protein synthesis and ubiquitination, potentially contributing to its anti-cancer effects. Moreover, the study revealed alterations in metabolic pathways upon ergosterol treatment, indicating its influence on metabolic processes critical for cancer development. This research sheds light on the multifaceted mechanisms through which ergosterol exerts anti-tumor effects, mainly focusing on Wnt/beta-catenin pathway modulation and metabolic pathway disruption. These findings provide valuable insights into the potential of ergosterol as a therapeutic candidate for breast cancer treatment, warranting further investigation and clinical application.
Collapse
Affiliation(s)
- Sunita Nilkhet
- Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Laboratory of Signal Transduction, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Wudtipong Vongthip
- Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Laboratory of Signal Transduction, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Pattawika Lertpatipanpong
- Laboratory of Signal Transduction, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Anchalee Prasansuklab
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tewin Tencomnao
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Siriporn Chuchawankul
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
- Immunomodulation of Natural Products Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Seung Joon Baek
- Laboratory of Signal Transduction, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
11
|
Li H, Yao Y, Hao R, Long C. Selective and effective suppression of pancreatic cancer through MNK inhibition. Immunopharmacol Immunotoxicol 2024:1-11. [PMID: 39138614 DOI: 10.1080/08923973.2024.2391462] [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: 01/05/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
Objective: The study aimed to explore the role of the Wnt/β-catenin signaling pathway in pancreatic cancer progression and chemoresistance, with a focus on identifying specific factors that distinguish between normal and tumor cells, thereby offering potential therapeutic targets. Materials and Methods: We analyzed levels of total and phosphorylated eukaryotic translation initiation factor 4E (eIF4E) and β-catenin in pancreatic cancer and normal pancreatic tissues. Functional assays were used to assess the impact of eIF4E phosphorylation on β-catenin signaling, cell proliferation, and chemoresistance, with MNK kinase involvement determined through gene depletion studies. The MNK kinase inhibitor eFT508 was evaluated for its effects on eIF4E phosphorylation, β-catenin activation, and cell viability in both in vitro and in vivo models of pancreatic cancer. Results: Both total and phosphorylated eIF4E, along with β-catenin, were significantly elevated in pancreatic cancer tissues compared to normal tissues. Phosphorylation of eIF4E at serine 209 was shown to activate β-catenin signaling, enhance cell proliferation, and contribute to chemoresistance in pancreatic cancer. Importantly, these effects were dependent on MNK kinase activity. Depletion of eIF4E reduced cell viability in both pancreatic cancer and normal cells, while depletion of MNK selectively decreased viability in pancreatic cancer cells. Treatment with eFT508 effectively inhibited eIF4E phosphorylation, suppressed β-catenin activation, and reduced pancreatic cancer cell growth and survival in vitro and in vivo, with minimal impact on normal cells. Conclusions: The MNK-eIF4E-β-catenin axis plays a critical role in pancreatic cancer progression and chemoresistance, distinguishing pancreatic cancer cells from normal cells. Targeting MNK kinases with inhibitors like eFT508 presents a promising therapeutic strategy for pancreatic cancer, with potential for selective efficacy and reduced toxicity.
Collapse
Affiliation(s)
- Hui Li
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Yang Yao
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Rui Hao
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Cheng Long
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| |
Collapse
|
12
|
Guo J, Jiang X, Lian J, Li H, Zhang F, Xie J, Deng J, Hou X, Du Z, Hao E. Evaluation of the effect of GSK-3β on liver cancer based on the PI3K/AKT pathway. Front Cell Dev Biol 2024; 12:1431423. [PMID: 39156976 PMCID: PMC11327086 DOI: 10.3389/fcell.2024.1431423] [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: 05/11/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
The PI3K/AKT/GSK-3β signaling pathway plays a pivotal role in numerous physiological and pathological processes, including cell proliferation, apoptosis, differentiation, and metabolic regulation. Aberrant activation of the PI3K/AKT pathway is intricately linked to development of tumor. GSK-3β, belonging to the serine/threonine protein kinase family, is crucial in the pathogenesis of liver cancer. As a key rate-limiting enzyme in the glucose metabolism pathway, GSK-3β significantly impacts the growth, proliferation, metastasis, and apoptosis of liver cancer cells. It is also implicated in chemotherapy resistance. Elevated expression of GSK-3β diminishes the sensitivity of liver cancer cells to chemotherapeutic agents, thereby playing a substantial role in the development of drug resistance. Consequently, targeting of GSK-3β, particularly within the PI3K/AKT signaling pathway, is regarded as a promising therapeutic strategy for liver cancer. The precise identification and subsequent modulation of this pathway represent a substantial potential for innovative clinical interventions in the management of liver cancer.
Collapse
Affiliation(s)
- Jiageng Guo
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Xinya Jiang
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jing Lian
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Huaying Li
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Fan Zhang
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Jinling Xie
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaotao Hou
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
13
|
Gancedo SN, Sahores A, Gómez N, Di Siervi N, May M, Yaneff A, de Sousa Serro MG, Fraunhoffer N, Dusetti N, Iovanna J, Shayo C, Davio CA, González B. The xenobiotic transporter ABCC4/MRP4 promotes epithelial mesenchymal transition in pancreatic cancer. Front Pharmacol 2024; 15:1432851. [PMID: 39114357 PMCID: PMC11303182 DOI: 10.3389/fphar.2024.1432851] [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: 05/14/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
The xenobiotic transporter ABCC4/MRP4 is highly expressed in pancreatic ductal adenocarcinoma (PDAC) and correlates with a more aggressive phenotype and metastatic propensity. Here, we show that ABCC4 promotes epithelial-mesenchymal transition (EMT) in PDAC, a hallmark process involving the acquisition of mesenchymal traits by epithelial cells, enhanced cell motility, and chemoresistance. Modulation of ABCC4 levels in PANC-1 and BxPC-3 cell lines resulted in the dysregulation of genes present in the EMT signature. Bioinformatic analysis on several cohorts including tumor samples, primary patient-derived cultured cells, patient-derived xenografts, and cell lines, revealed a positive correlation between ABCC4 expression and EMT markers. We also characterized the ABCC4 cistrome and identified four candidate clusters in the distal promoter and intron one that showed differential binding of pro-epithelial FOXA1 and pro-mesenchymal GATA2 transcription factors in low ABCC4-expressing HPAF-II and high ABCC4-expressing PANC-1 xenografts. HPAF-II xenografts showed exclusive binding of FOXA1, and PANC-1 xenografts exclusive binding of GATA2, at ABCC4 clusters, consistent with their low and high EMT phenotype respectively. Our results underscore ABCC4/MRP4 as a valuable prognostic marker and a potential therapeutic target to treat PDAC subtypes with prominent EMT features, such as the basal-like/squamous subtype, characterized by worse prognosis and no effective therapies.
Collapse
Affiliation(s)
- S. N. Gancedo
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - A. Sahores
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Programa Franco-argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
| | - N. Gómez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - N. Di Siervi
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - M. May
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - A. Yaneff
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Programa Franco-argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
| | - M. G. de Sousa Serro
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - N. Fraunhoffer
- Programa Franco-argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, CNRS UMR, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
- Equipe Labellisée La Ligue, Marseille, France
| | - N. Dusetti
- Programa Franco-argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, CNRS UMR, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
- Equipe Labellisée La Ligue, Marseille, France
| | - J. Iovanna
- Programa Franco-argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, CNRS UMR, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
- Equipe Labellisée La Ligue, Marseille, France
- Hospital de Alta Complejidad El Cruce, Argentina. Universidad Nacional Arturo Jauretche, Buenos Aires, Argentina
| | - C. Shayo
- Instituto de Biología y Medicina Experimental (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - C. A. Davio
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Programa Franco-argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
| | - B. González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- Programa Franco-argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
| |
Collapse
|
14
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
15
|
Golivi Y, Kumari S, Farran B, Alam A, Peela S, Nagaraju GP. Small molecular inhibitors: Therapeutic strategies for pancreatic cancer. Drug Discov Today 2024; 29:104053. [PMID: 38849028 DOI: 10.1016/j.drudis.2024.104053] [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: 03/30/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Pancreatic cancer (PC), a disease with high heterogeneity and a dense stromal microenvironment, presents significant challenges and a bleak prognosis. Recent breakthroughs have illuminated the crucial interplay among RAS, epidermal growth factor receptor (EGFR), and hedgehog pathways in PC progression. Small molecular inhibitors have emerged as a potential solution with their advantages of oral administration and the ability to target intracellular and extracellular sites effectively. However, despite the US FDA approving over 100 small-molecule targeted antitumor drugs, challenges such as low response rates and drug resistance persist. This review delves into the possibility of using small molecules to treat persistent or spreading PC, highlighting the challenges and the urgent need for a diverse selection of inhibitors to develop more effective treatment strategies.
Collapse
Affiliation(s)
- Yuvasri Golivi
- Department of Bioscience and Biotechnology, Banasthali University, Banasthali, RJ 304 022, India
| | - Seema Kumari
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GIS, GITAM, Visakhapatnam, Andhra Pradesh 530045, India
| | - Batoul Farran
- Department of Hematology and Oncology, Henry Ford Health, Detroit, MI 48202, USA
| | - Afroz Alam
- Department of Bioscience and Biotechnology, Banasthali University, Banasthali, RJ 304 022, India
| | - Sujatha Peela
- Department of Biotechnology, Dr. B. R. Ambedkar University, Srikakulam, Andhra Pradesh, 532001, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
16
|
Ma Y, Lv H, Xing F, Xiang W, Wu Z, Feng Q, Wang H, Yang W. Cancer stem cell-immune cell crosstalk in the tumor microenvironment for liver cancer progression. Front Med 2024; 18:430-445. [PMID: 38600350 DOI: 10.1007/s11684-023-1049-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/15/2023] [Indexed: 04/12/2024]
Abstract
Crosstalk between cancer cells and the immune microenvironment is determinant for liver cancer progression. A tumor subpopulation called liver cancer stem cells (CSCs) significantly accounts for the initiation, metastasis, therapeutic resistance, and recurrence of liver cancer. Emerging evidence demonstrates that the interaction between liver CSCs and immune cells plays a crucial role in shaping an immunosuppressive microenvironment and determining immunotherapy responses. This review sheds light on the bidirectional crosstalk between liver CSCs and immune cells for liver cancer progression, as well as the underlying molecular mechanisms after presenting an overview of liver CSCs characteristic and their microenvironment. Finally, we discuss the potential application of liver CSCs-targeted immunotherapy for liver cancer treatment.
Collapse
Affiliation(s)
- Yue Ma
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Hongwei Lv
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Fuxue Xing
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Wei Xiang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Zixin Wu
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Qiyu Feng
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China
| | - Hongyang Wang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Wen Yang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China.
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China.
- Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai, 200438, China.
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| |
Collapse
|
17
|
Kongsintaweesuk S, Klungsaeng S, Intuyod K, Techasen A, Pairojkul C, Luvira V, Pinlaor S, Pinlaor P. Microcystin-leucine arginine induces the proliferation of cholangiocytes and cholangiocarcinoma cells through the activation of the Wnt/β-catenin signaling pathway. Heliyon 2024; 10:e30104. [PMID: 38720699 PMCID: PMC11076882 DOI: 10.1016/j.heliyon.2024.e30104] [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: 01/15/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
Background Microcystin-leucine arginine (MC-LR) is a cyanobacterial hepatotoxic toxin found in water sources worldwide, including in northeastern Thailand, where opisthorchiasis-associated cholangiocarcinoma (CCA) is most prevalent. MC-LR is a potential carcinogen; however, its involvement in liver fluke-associated CCA remains ambiguous. Here, we aimed to evaluate the effect of MC-LR on the progression of CCA via the Wnt/β-catenin pathway in vitro. Methods Cell division, migration, cell cycle transition, and MC-LR transporter expression were evaluated in vitro through MTT assay, wound healing assay, flow cytometry, and immunofluorescence staining, respectively. Following a 24-h treatment of cultured cells with 1, 10, 100, and 1,000 nM of MC-LR, the proliferative effect of MC-LR on the Wnt/β-catenin signaling pathway was investigated using immunoblotting and qRT-PCR analysis. Immunohistochemistry was used to determine β-catenin expression in CCA tissue compared to adjacent tissue. Results Human immortalized cholangiocyte cells (MMNK-1) and a human cell line established from opisthorchiasis-associated CCA (KKU-213B) expressed the MC-LR transporter and internalized MC-LR. Exposure to 10 nM and 100 nM of MC-LR notably enhanced cells division and migration in both cell lines (P < 0.05) and markedly elevated the percentage of S phase cells (P < 0.05). MC-LR elevated PP2A expression by activating the Wnt/β-catenin signaling pathway and suppressing phosphatase activity. Inhibition of the β-catenin destruction complex genes (Axin1 and APC) led to the upregulation of β-catenin and its downstream target genes (Cyclin D1 and c-Jun). Inhibition of Wnt/β-catenin signaling by MSAB confirmed these results. Additionally, β-catenin was significantly expressed in cancerous tissue compared to adjacent areas (P < 0.001). Conclusions Our findings suggest that MC-LR promotes cell proliferation and progression of CCA through Wnt/β-catenin pathway. Further evaluation using invivo experiments is needed to confirm this observation. This finding could promote health awareness regarding MC-LR intake and risk of CCA.
Collapse
Affiliation(s)
- Suppakrit Kongsintaweesuk
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- Medical Sciences Program, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sirinapha Klungsaeng
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Kitti Intuyod
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Anchalee Techasen
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- School of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Vor Luvira
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Somchai Pinlaor
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Porntip Pinlaor
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- School of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
18
|
Huang YP, Yeh CA, Ma YS, Chen PY, Lai KC, Lien JC, Hsieh WT. PW06 suppresses cancer cell metastasis in human pancreatic carcinoma MIA PaCa-2 cells via the inhibitions of p-Akt/mTOR/NF-κB and MMP2/MMP9 signaling pathways in vitro. ENVIRONMENTAL TOXICOLOGY 2024; 39:2768-2781. [PMID: 38264921 DOI: 10.1002/tox.24143] [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: 06/29/2023] [Revised: 12/14/2023] [Accepted: 01/06/2024] [Indexed: 01/25/2024]
Abstract
PW06 [(E)-3-(9-ethyl-9H-carbazol-3-yl)-1-(2,5-dimethoxyphenyl) prop-2-en-1-one], a kind of the carbazole derivative containing chalcone moiety, induced cell apoptosis in human pancreatic carcinoma in vitro. There is no investigation to show that PW06 inhibits cancer cell metastasis in human pancreatic carcinoma in vitro. Herein, PW06 (0.1-0.8 μM) significantly exists in the antimetastatic activities of human pancreatic carcinoma MIA PaCa-2 cells in vitro. Wound healing assay shows PW06 at 0.2 μM suppressed cell mobility by 7.45 and 16.55% at 6 and 24 hours of treatments. PW06 at 0.1 and 0.2 μM reduced cell mobility by 14.72 and 21.8% for 48 hours of treatment. Transwell chamber assay indicated PW06 (0.1-0.2 μM) suppressed the cell migration (decreased 26.67-35.42%) and invasion (decreased 48.51-68.66%). Atomic force microscopy assay shows PW06 (0.2 μM) significantly changed the shape of cell morphology. The gelatin zymography assay indicates PW06 decreased MMP2's and MMP9's activities at 48 hours of treatment. Western blotting assay further confirms PW06 reduced levels of MMP2 and MMP9 and increased protein expressions of EGFR, SOS1, and Ras. PW06 also increased the p-JNK, p-ERK, and p-p38. PW06 increased the expression of PI3K, PTEN, Akt, GSK3α/β, and E-cadherin. Nevertheless, results also show PW06 decreased p-Akt, mTOR, NF-κB, p-GSK3β, β-catenin, Snail, N-cadherin, and vimentin in MIA PaCa-2 cells. The confocal laser microscopy examination shows PW06 increased E-cadherin but decreased vimentin in MIA PaCa-2 cells. Together, our findings strongly suggest that PW06 inhibited the p-Akt/mTOR/NF-κB/MMPs pathways, increased E-cadherin, and decreased N-cadherin/vimentin, suppressing the migration and invasion in MIA PaCa-2 cells in vitro.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-An Yeh
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Chinese Medicine, E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Po-Yuan Chen
- Department of Biological Science and Technology, College of Life Science, China Medical University, Taichung, Taiwan
| | - Kuang-Chi Lai
- Department of Medical Laboratory Science and Biotechnology, College of Medical Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan
- Department of Surgery, School of Medicine, China Medical University, Taichung, Taiwan
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Wen-Tsong Hsieh
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Pharmacology, China Medical University, Taichung, Taiwan
| |
Collapse
|
19
|
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
|
20
|
Takata T, Nakamura A, Yasuda H, Miyake H, Sogame Y, Sawai Y, Hayakawa M, Mochizuki K, Nakao R, Ogata T, Ikoma H, Konishi E, Harada Y, Otsuji E, Itoh Y, Tanaka H. Pathophysiological Implications of Protein Lactylation in Pancreatic Epithelial Tumors. Acta Histochem Cytochem 2024; 57:57-66. [PMID: 38695038 PMCID: PMC11058462 DOI: 10.1267/ahc.24-00010] [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: 01/19/2024] [Accepted: 02/02/2024] [Indexed: 05/04/2024] Open
Abstract
Protein lactylation is a post-translational modification associated with glycolysis. Although recent evidence indicates that protein lactylation is involved in epigenetic gene regulation, its pathophysiological significance remains unclear, particularly in neoplasms. Herein, we investigated the potential involvement of protein lactylation in the molecular mechanisms underlying benign and malignant pancreatic epithelial tumors, as well as its role in the response of pancreatic cancer (PC) cells to gemcitabine. Increased lactylation was observed in the nuclei of intraductal papillary mucinous adenoma, non-invasive intraductal papillary mucinous carcinoma, and invasive carcinoma, in parallel to the upregulation of hypoxia-inducible factor-1α. This observation indicated that a hypoxia-associated increase in nuclear protein lactylation could be a biochemical hallmark in pancreatic epithelial tumors. The standard PC chemotherapy drug gemcitabine suppressed histone lactylation in vitro, suggesting that histone lactylation might be relevant to its mechanism of action. Taken together, our findings suggest that protein lactylation may be involved in the development of pancreatic epithelial tumors and could represent a potential therapeutic target for PC.
Collapse
Affiliation(s)
- Tomoki Takata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akihiro Nakamura
- Central Research Facility, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hayato Miyake
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshio Sogame
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuki Sawai
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michiyo Hayakawa
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kentaro Mochizuki
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryuta Nakao
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
21
|
Malla R, Viswanathan S, Makena S, Kapoor S, Verma D, Raju AA, Dunna M, Muniraj N. Revitalizing Cancer Treatment: Exploring the Role of Drug Repurposing. Cancers (Basel) 2024; 16:1463. [PMID: 38672545 PMCID: PMC11048531 DOI: 10.3390/cancers16081463] [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: 03/02/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer persists as a global challenge necessitating continual innovation in treatment strategies. Despite significant advancements in comprehending the disease, cancer remains a leading cause of mortality worldwide, exerting substantial economic burdens on healthcare systems and societies. The emergence of drug resistance further complicates therapeutic efficacy, underscoring the urgent need for alternative approaches. Drug repurposing, characterized by the utilization of existing drugs for novel clinical applications, emerges as a promising avenue for addressing these challenges. Repurposed drugs, comprising FDA-approved (in other disease indications), generic, off-patent, and failed medications, offer distinct advantages including established safety profiles, cost-effectiveness, and expedited development timelines compared to novel drug discovery processes. Various methodologies, such as knowledge-based analyses, drug-centric strategies, and computational approaches, play pivotal roles in identifying potential candidates for repurposing. However, despite the promise of repurposed drugs, drug repositioning confronts formidable obstacles. Patenting issues, financial constraints associated with conducting extensive clinical trials, and the necessity for combination therapies to overcome the limitations of monotherapy pose significant challenges. This review provides an in-depth exploration of drug repurposing, covering a diverse array of approaches including experimental, re-engineering protein, nanotechnology, and computational methods. Each of these avenues presents distinct opportunities and obstacles in the pursuit of identifying novel clinical uses for established drugs. By examining the multifaceted landscape of drug repurposing, this review aims to offer comprehensive insights into its potential to transform cancer therapeutics.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Sathiyapriya Viswanathan
- Department of Biochemistry, ACS Medical College and Hospital, Chennai 600007, Tamil Nadu, India;
| | - Sree Makena
- Maharajah’s Institute of Medical Sciences and Hospital, Vizianagaram 535217, Andhra Pradesh, India
| | - Shruti Kapoor
- Department of Genetics, University of Alabama, Birmingham, AL 35233, USA
| | - Deepak Verma
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | - Manikantha Dunna
- Center for Biotechnology, Jawaharlal Nehru Technological University, Hyderabad 500085, Telangana, India
| | - Nethaji Muniraj
- Center for Cancer and Immunology Research, Children’s National Hospital, 111, Michigan Ave NW, Washington, DC 20010, USA
| |
Collapse
|
22
|
Katoozian F, Abedi Kichi Z, Sharifi R, Shirvani-Farsani Z. The Expression Analysis of Long Non-coding RNAs Related to Wnt/β-Catenin Signaling in Pancreatic Cancer Patients. Biochem Genet 2024:10.1007/s10528-024-10779-5. [PMID: 38594570 DOI: 10.1007/s10528-024-10779-5] [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: 01/22/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024]
Abstract
Background The oncogenic Wnt/β-catenin signaling plays a critical role in carcinogenesis, prognosis, and resistance to therapy. Pancreatic cancer (PC) has high mortality because of its poor prognosis. Several studies have suggested that lncRNAs are directly involved in the development and progression of PC as well as in Wnt/β-catenin signaling. In this study, we investigated and compared the expression of Wnt/β-catenin signaling-related ZFAS1 and HCG11 lncRNAs, and their targets, CTNNB1 and IGF2BP1 genes in the blood of patients with PC and healthy individuals. A total of 47 PC patients and 50 healthy individuals participated in this study. RNA was extracted from the peripheral blood samples of participants, and cDNA was synthesized. The expression level of the selected genes was quantified by real-time PCR. The expression of HCG11 lncRNA and CTNNB1 genes in patients with PC was significantly upregulated compared to healthy individuals, and the expression of the ZFAS1 lncRNA was significantly downregulated. According to the analysis of the ROC curve, the diagnostic powers of ZFAS1 and CTNNB1 in PC were 0.67 and 0.69, respectively. Altogether, the present study suggests a role for ZFAS1 and HCG11 lncRNAs and CTNNB1 and IGF2BP1 in the pathogenesis of pancreatic cancer. Moreover, the peripheral expression of these lncRNAs may be useful as potential biomarkers for PC.
Collapse
Affiliation(s)
- Fatemeh Katoozian
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Zahra Abedi Kichi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany
| | - Roya Sharifi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Zeinab Shirvani-Farsani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
23
|
Jiménez DJ, Javed A, Rubio-Tomás T, Seye-Loum N, Barceló C. Clinical and Preclinical Targeting of Oncogenic Pathways in PDAC: Targeted Therapeutic Approaches for the Deadliest Cancer. Int J Mol Sci 2024; 25:2860. [PMID: 38474109 DOI: 10.3390/ijms25052860] [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: 12/21/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 03/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related death worldwide. It is commonly diagnosed in advanced stages and therapeutic interventions are typically constrained to systemic chemotherapy, which yields only modest clinical outcomes. In this review, we examine recent developments in targeted therapy tailored to address distinct molecular pathway alteration required for PDAC. Our review delineates the principal signaling pathways and molecular mechanisms implicated in the initiation and progression of PDAC. Subsequently, we provide an overview of prevailing guidelines, ongoing investigations, and prospective research trajectories related to targeted therapeutic interventions, drawing insights from randomized clinical trials and other pertinent studies. This review focus on a comprehensive examination of preclinical and clinical data substantiating the efficacy of these therapeutic modalities, emphasizing the potential of combinatorial regimens and novel therapies to enhance the quality of life for individuals afflicted with PDAC. Lastly, the review delves into the contemporary application and ongoing research endeavors concerning targeted therapy for PDAC. This synthesis serves to bridge the molecular elucidation of PDAC with its clinical implications, the evolution of innovative therapeutic strategies, and the changing landscape of treatment approaches.
Collapse
Affiliation(s)
- Diego J Jiménez
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Aadil Javed
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Teresa Rubio-Tomás
- School of Medicine, University of Crete, 70013 Herakleion, Crete, Greece
| | - Ndioba Seye-Loum
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Carles Barceló
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| |
Collapse
|
24
|
Pramanik N, Gupta A, Ghanwatkar Y, Mahato RI. Recent advances in drug delivery and targeting for the treatment of pancreatic cancer. J Control Release 2024; 366:231-260. [PMID: 38171473 PMCID: PMC10922996 DOI: 10.1016/j.jconrel.2023.12.053] [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: 09/17/2023] [Revised: 11/24/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Despite significant treatment efforts, pancreatic ductal adenocarcinoma (PDAC), the deadliest solid tumor, is still incurable in the preclinical stages due to multifacet stroma, dense desmoplasia, and immune regression. Additionally, tumor heterogeneity and metabolic changes are linked to low grade clinical translational outcomes, which has prompted the investigation of the mechanisms underlying chemoresistance and the creation of effective treatment approaches by selectively targeting genetic pathways. Since targeting upstream molecules in first-line oncogenic signaling pathways typically has little clinical impact, downstream signaling pathways have instead been targeted in both preclinical and clinical studies. In this review, we discuss how the complexity of various tumor microenvironment (TME) components and the oncogenic signaling pathways that they are connected to actively contribute to the development and spread of PDAC, as well as the ways that recent therapeutic approaches have been targeted to restore it. We also illustrate how many endogenous stimuli-responsive linker-based nanocarriers have recently been developed for the specific targeting of distinct oncogenes and their downstream signaling cascades as well as their ongoing clinical trials. We also discuss the present challenges, prospects, and difficulties in the development of first-line oncogene-targeting medicines for the treatment of pancreatic cancer patients.
Collapse
Affiliation(s)
- Nilkamal Pramanik
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aditya Gupta
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yashwardhan Ghanwatkar
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
25
|
Aliakbarian M, Ferns GA, Shabestari MM, Ahmadzadeh AM, Abdollahzade A, Rahimi H, Khodashahi R, Arjmand MH. Elucidating the Role of Pro-renin Receptors in Pancreatic Ductal Adenocarcinoma Progression: A Novel Therapeutic Target in Cancer Therapy. Curr Cancer Drug Targets 2024; 24:881-889. [PMID: 38279719 DOI: 10.2174/0115680096279288231205105904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 01/28/2024]
Abstract
Pancreatic cancer is a highly aggressive malignancy with a very poor prognosis. The 5- year survival in these patients is very low, and most patients develop drug resistance to current therapies, so additional studies are needed to identify the potential role of new drug targets for the treatment of pancreatic cancer. Recent investigations have been performed regarding the roles of pro-renin receptors (PRR) in the initiation and development of cancers. PRR is a component of the local renin-angiotensin system (RAS). Local tissue RAS has been known in diverse organ systems, including the pancreas. Various investigations have implicated that PRRs are associated with the upregulation of various signaling pathways, like the renin-angiotensin system pathway, PI3K/Akt/mTOR, and the Wnt-signaling pathways, to contribute to pathological conditions, including cancer. In this review, we presented an overview of the role of PRR in the progression of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Mohsen Aliakbarian
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Department of Biochemistry, Division of Medical, Brighton & Sussex Medical School, Brighton, UK
| | | | - Amir Mahmoud Ahmadzadeh
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Radiology, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aref Abdollahzade
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hoda Rahimi
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rozita Khodashahi
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad-Hassan Arjmand
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Lv S, Zhang J, Peng X, Liu H, Liu Y, Wei F. Ubiquitin signaling in pancreatic ductal adenocarcinoma. Front Mol Biosci 2023; 10:1304639. [PMID: 38174069 PMCID: PMC10761520 DOI: 10.3389/fmolb.2023.1304639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor of the digestive system, characterized by rapid progression and being prone to metastasis. Few effective treatment options are available for PDAC, and its 5-year survival rate is less than 9%. Many cell biological and signaling events are involved in the development of PDAC, among which protein post-translational modifications (PTMs), such as ubiquitination, play crucial roles. Catalyzed mostly by a three-enzyme cascade, ubiquitination induces changes in protein activity mainly by altering their stability in PDAC. Due to their role in substrate recognition, E3 ubiquitin ligases (E3s) dictate the outcome of the modification. Ubiquitination can be reversed by deubiquitylases (DUBs), which, in return, modified proteins to their native form. Dysregulation of E3s or DUBs that disrupt protein homeostasis is involved in PDAC. Moreover, the ubiquitination system has been exploited to develop therapeutic strategies, such as proteolysis-targeting chimeras (PROTACs). In this review, we summarize recent progress in our understanding of the role of ubiquitination in the development of PDAC and offer perspectives in the design of new therapies against this highly challenging disease.
Collapse
Affiliation(s)
- Shengnan Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyu Peng
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huan Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yan Liu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
27
|
Fernandes Q, Therachiyil L, Khan AQ, Bedhiafi T, Korashy HM, Bhat AA, Uddin S. Shrinking the battlefield in cancer therapy: Nanotechnology against cancer stem cells. Eur J Pharm Sci 2023; 191:106586. [PMID: 37729956 DOI: 10.1016/j.ejps.2023.106586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
Cancer remains one of the leading causes of mortality worldwide, presenting a significant healthcare challenge owing to the limited efficacy of current treatments. The application of nanotechnology in cancer treatment leverages the unique optical, magnetic, and electrical attributes of nanomaterials to engineer innovative, targeted therapies. Specifically, manipulating nanomaterials allows for enhanced drug loading efficiency, improved bioavailability, and targeted delivery systems, reducing the non-specific cytotoxic effects characteristic of conventional chemotherapies. Furthermore, recent advances in nanotechnology have demonstrated encouraging results in specifically targeting CSCs, a key development considering the role of these cells in disease recurrence and resistance to treatment. Despite these breakthroughs, the clinical approval rates of nano-drugs have not kept pace with research advances, pointing to existing obstacles that must be addressed. In conclusion, nanotechnology presents a novel, powerful tool in the fight against cancer, particularly in targeting the elusive and treatment-resistant CSCs. This comprehensive review delves into the intricacies of nanotherapy, explicitly targeting cancer stem cells, their markers, and associated signaling pathways.
Collapse
Affiliation(s)
- Queenie Fernandes
- College of Medicine, Qatar University, Doha, Qatar; Translational Cancer Research Facility, Hamad Medical Corporation, National Center for Cancer Care and Research, PO. Box 3050, Doha, Qatar
| | - Lubna Therachiyil
- Academic Health System, Hamad Medical Corporation, Translational Research Institute, Doha 3050, Qatar; Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Abdul Q Khan
- Academic Health System, Hamad Medical Corporation, Translational Research Institute, Doha 3050, Qatar
| | - Takwa Bedhiafi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Shahab Uddin
- College of Medicine, Qatar University, Doha, Qatar; Academic Health System, Hamad Medical Corporation, Dermatology Institute, Doha 3050, Qatar; Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar; Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 22602, India.
| |
Collapse
|
28
|
Elkady N, Aldesoky AI, Allam DM. Can β-catenin, Tenascin and Fascin be potential biomarkers for personalized therapy in Gastric carcinoma? J Immunoassay Immunochem 2023; 44:396-417. [PMID: 37694977 DOI: 10.1080/15321819.2023.2251564] [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] [Indexed: 09/12/2023]
Abstract
Gastric carcinoma (GC) is one of the most prevalent cancers worldwide and the fourth leading cause of cancer-related death. Studying the molecular profile of GC is essential for developing targeted therapies. β-catenin, Tenascin, and Fascin expression are among the molecular abnormalities that are claimed to cause GC progression and chemoresistance. Therefore, they could be used as potential therapeutic targets. This study aimed to evaluate β-catenin, Tenascin, and Fascin expression and their possible roles as prognostic and predictive biomarkers in GC using immunohistochemistry. This retrospective study included 84 GC cases. Tissue microarrays were constructed, followed by β-catenin, Tenascin, and Fascin immunostaining. Their expression was assessed and compared with clinicopathological parameters and survival data. The study results revealed that β-catenin nucleocytoplasmic expression, positive Tenascin, and Fascin expressions were detected in 86.9%, 70%, and 59.5% of cases, respectively. Their expression was significantly associated with poor prognostic parameters, such as deeper tumor invasion, lymph node metastasis, advanced pathological stage, vascular invasion, positive omental nodules, poor response to chemotherapy, and short overall survival. Hence, nucleocytoplasmic β-catenin expression together with Tenascin and Fascin positivity can be potential prognostic and predictive markers, and they can be used as therapeutic targets for GC.
Collapse
Affiliation(s)
- Noha Elkady
- Pathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Amira I Aldesoky
- Clinical oncology and nuclear medicine department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Dina Mohamed Allam
- Pathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| |
Collapse
|
29
|
Kashiwagi R, Funayama R, Aoki S, Matsui A, Klein S, Sato Y, Suzuki T, Murakami K, Inoue K, Iseki M, Masuda K, Mizuma M, Naito H, Duda DG, Unno M, Nakayama K. Collagen XVII regulates tumor growth in pancreatic cancer through interaction with the tumor microenvironment. Cancer Sci 2023; 114:4286-4298. [PMID: 37688308 PMCID: PMC10637054 DOI: 10.1111/cas.15952] [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: 04/01/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Expression of the gene for collagen XVII (COL17A1) in tumor tissue is positively or negatively associated with patient survival depending on cancer type. High COL17A1 expression is thus a favorable prognostic marker for breast cancer but unfavorable for pancreatic cancer. This study explored the effects of COL17A1 expression on pancreatic tumor growth and their underlying mechanisms. Analysis of published single-cell RNA-sequencing data for human pancreatic cancer tissue revealed that COL17A1 was expressed predominantly in cancer cells rather than surrounding stromal cells. Forced expression of COL17A1 did not substantially affect the proliferation rate of the mouse pancreatic cancer cell lines KPC and AK4.4 in vitro. However, in mouse homograft tumor models in which KPC or AK4.4 cells were injected into syngeneic C57BL/6 or FVB mice, respectively, COL17A1 expression promoted or suppressed tumor growth, respectively, suggesting that the effect of COL17A1 on tumor growth was influenced by the tumor microenvironment. RNA-sequencing analysis of tumor tissue revealed effects of COL17A1 on gene expression profiles (including the expression of genes related to cell proliferation, the immune response, Wnt signaling, and Hippo signaling) that differed between C57BL/6-KPC and FVB-AK4.4 tumors. Our data thus suggest that COL17A1 promotes or suppresses cancer progression in a manner dependent on the interaction of tumor cells with the tumor microenvironment.
Collapse
Affiliation(s)
- Ryosuke Kashiwagi
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Ryo Funayama
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Shuichi Aoki
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Aya Matsui
- Department of Vascular Physiology, Graduate School of Medical ScienceKanazawa UniversityKanazawaJapan
| | - Sebastian Klein
- PathologyUniversity Hospital CologneCologneGermany
- Radiation Oncology/Steele Laboratories for Tumor BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Yukihiro Sato
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Tsubasa Suzuki
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Keigo Murakami
- Department of Investigative Pathology, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Koetsu Inoue
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Masahiro Iseki
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Kunihiro Masuda
- Department of SurgerySouth Miyagi Medical CenterShibata‐gunJapan
| | - Masamichi Mizuma
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Hisamichi Naito
- Department of Vascular Physiology, Graduate School of Medical ScienceKanazawa UniversityKanazawaJapan
| | - Dan G. Duda
- Radiation Oncology/Steele Laboratories for Tumor BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Michiaki Unno
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Keiko Nakayama
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
| |
Collapse
|
30
|
Itzhak I, Bareket-Samish A, Fishman P. Namodenoson Inhibits the Growth of Pancreatic Carcinoma via Deregulation of the Wnt/β-catenin, NF-κB, and RAS Signaling Pathways. Biomolecules 2023; 13:1584. [PMID: 38002266 PMCID: PMC10669398 DOI: 10.3390/biom13111584] [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: 09/28/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Namodenoson, an A3 adenosine receptor (A3AR) agonist, is currently being used in a phase III trial in advanced liver cancer. We examined the anti-growth effect of namodenoson on pancreatic carcinoma cells and investigated the molecular mechanism involved. BxPC-3 pancreatic carcinoma cells were cultured with namodenoson (5-20 nM for 24 h at 37 °C), and the Presto Blue assay was used to monitor cell growth. Western blot analyses were performed on BxPC-3 cells (20 nM namodenoson for 24 h at 37 °C) to evaluate the expression levels of cell growth regulatory proteins. In vivo studies involved the subcutaneous inoculation of BxPC-3 cells into nude mice, randomizing the mice into namodenoson (10 μg/kg twice daily for 35 days) vs. control, and monitoring tumor size twice weekly. Treatment with namodenoson was associated with the significant dose-dependent inhibition of BxPC-3 cell growth, which was mitigated by the A3AR antagonist MRS1523. Western blot analyses showed that namodenoson treatment modulated the expression of NF-κB, as well as proteins in the Wnt/β-catenin and the RAS signaling pathways, leading to the upregulation of apoptotic proteins (Bad, Bax). In vivo studies also showed the significant inhibition of pancreatic carcinoma tumor growth with namodenoson. In conclusion, our findings support the continued development of namodenoson as a treatment for pancreatic cancer.
Collapse
|
31
|
Huang L, Liu Y, Pan Y, Liu C, Gao H, Ren Q, Wang J, Wang H, Zhang Y, Wu A. Elaiophylin Elicits Robust Anti-Tumor Responses via Apoptosis Induction and Attenuation of Proliferation, Migration, Invasion, and Angiogenesis in Pancreatic Cancer Cells. Molecules 2023; 28:7205. [PMID: 37894684 PMCID: PMC10608934 DOI: 10.3390/molecules28207205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic cancer remains a formidable challenge in oncology due to its aggressive nature and limited treatment options. In this study, we investigate the potential therapeutic efficacy of elaiophylin, a novel compound, in targeting BxPC-3 and PANC-1 pancreatic cancer cells. We comprehensively explore elaiophylin's impact on apoptosis induction, proliferation inhibition, migration suppression, invasion attenuation, and angiogenesis inhibition, key processes contributing to cancer progression and metastasis. The results demonstrate that elaiophylin exerts potent pro-apoptotic effects, inducing a substantial increase in apoptotic cells. Additionally, elaiophylin significantly inhibits proliferation, migration, and invasion of BxPC-3 and PANC-1 cells. Furthermore, elaiophylin exhibits remarkable anti-angiogenic activity, effectively disrupting tube formation in HUVECs. Moreover, elaiophylin significantly inhibits the Wnt/β-Catenin signaling pathway. Our findings collectively demonstrate the multifaceted potential of elaiophylin as a promising therapeutic agent against pancreatic cancer via inhibition of the Wnt/β-Catenin signaling pathway. By targeting diverse cellular processes crucial for cancer progression, elaiophylin emerges as a prospective candidate for future targeted therapies. Further investigation of the in vivo efficacy of elaiophylin is warranted, potentially paving the way for novel and effective treatment approaches in pancreatic cancer management.
Collapse
Affiliation(s)
- Lufen Huang
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Yufeng Liu
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Yiru Pan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China;
| | - Chao Liu
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Huijie Gao
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Qiang Ren
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Jianan Wang
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Huiyun Wang
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Yuntao Zhang
- Department of Pharmacy, Jining Medical University, Rizhao 276500, China; (L.H.); (Y.L.); (C.L.); (H.G.); (Q.R.); (J.W.); (H.W.)
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China;
| |
Collapse
|
32
|
Samant C, Kale R, Bokare A, Verma M, Pai KSR, Bhonde M. PAK4 inhibition significantly potentiates Gemcitabine activity in PDAC cells via inhibition of Wnt/β-catenin, p-ERK/MAPK and p-AKT/PI3K pathways. Biochem Biophys Rep 2023; 35:101544. [PMID: 37720313 PMCID: PMC10500449 DOI: 10.1016/j.bbrep.2023.101544] [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: 06/25/2023] [Revised: 08/21/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) remains one of the most difficult to treat cancers. Gemcitabine is still the standard of care treatment for PDAC but with modest survival benefit and well reported resistance. Here we explored potential of inhibiting p21 activated kinase 4 (PAK4), a downstream protein of KRAS oncogenic pathway, in combination with Gemcitabine in PDAC cells. PAK4 inhibition by KPT-9274 led to significant potentiation of Gemcitabine activity in PDAC cells, with an increase in apoptosis, DNA damage and cell cycle arrest. At molecular level, PAK4 inhibition dose dependently inhibited Gemcitabine-induced β-catenin, c-JUN and Ribonucleotide Reductase subunit 2 (RRM2) levels. PAK4 inhibition further inhibited levels of phosphorylated ERK (p-ERK); Gemcitabine-induced phosphorylated AKT (p-AKT), phosphorylated and total c-Myc. These results suggest possible role of β-catenin, p-ERK and p-AKT, key effector proteins of Wnt/β-catenin, MAPK and PI3K pathways respectively, in sensitisation of Gemcitabine activity with PAK4 inhibition. Our data unravel probable molecular mechanisms behind combination of PAK4 inhibition with Gemcitabine to counter PDAC, which may be unequivocally proved further with knock down of PAK4. Our findings provide a strong rationale to exploit the combination therapy of Gemcitabine and PAK4 inhibitor for PDAC at pre-clinical and clinical levels.
Collapse
Affiliation(s)
- Charudatt Samant
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| | - Ramesh Kale
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| | - Anand Bokare
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| | - Mahip Verma
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| | - K. Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Mandar Bhonde
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| |
Collapse
|
33
|
Sahni S, Nahm C, Ahadi MS, Sioson L, Byeon S, Chou A, Maloney S, Moon E, Pavlakis N, Gill AJ, Samra J, Mittal A. Gene expression profiling of pancreatic ductal adenocarcinomas in response to neoadjuvant chemotherapy. Cancer Med 2023; 12:18050-18061. [PMID: 37533202 PMCID: PMC10523964 DOI: 10.1002/cam4.6411] [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: 03/27/2023] [Revised: 07/09/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
AIM Pancreatic ductal adenocarcinoma (PDAC) has the lowest survival rate of all major cancers. Chemotherapy is the mainstay systemic therapy for PDAC, and chemoresistance is a major clinical problem leading to therapeutic failure. This study aimed to identify key differences in gene expression profile in tumors from chemoresponsive and chemoresistant patients. METHODS Archived formalin-fixed paraffin-embedded tumor tissue samples from patients treated with neoadjuvant chemotherapy were obtained during surgical resection. Specimens were macrodissected and gene expression analysis was performed. Multi- and univariate statistical analysis was performed to identify differential gene expression profile of tumors from good (0%-30% residual viable tumor [RVT]) and poor (>30% RVT) chemotherapy-responders. RESULTS Initially, unsupervised multivariate modeling was performed by principal component analysis, which demonstrated a distinct gene expression profile between good- and poor-chemotherapy responders. There were 396 genes that were significantly (p < 0.05) downregulated (200 genes) or upregulated (196 genes) in tumors from good responders compared to poor responders. Further supervised multivariate analysis of significant genes by partial least square (PLS) demonstrated a highly distinct gene expression profile between good- and poor responders. A gene biomarker of panel (IL18, SPA17, CD58, PTTG1, MTBP, ABL1, SFRP1, CHRDL1, IGF1, and CFD) was selected based on PLS model, and univariate regression analysis of individual genes was performed. The identified biomarker panel demonstrated a very high ability to diagnose good-responding PDAC patients (AUROC: 0.977, sensitivity: 82.4%; specificity: 87.0%). CONCLUSION A distinct tumor biological profile between PDAC patients who either respond or not respond to chemotherapy was identified.
Collapse
Affiliation(s)
- Sumit Sahni
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
| | - Christopher Nahm
- Western Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
| | - Mahsa S. Ahadi
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Department of Anatomical Pathology, NSW Health PathologyRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - Loretta Sioson
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Department of Anatomical Pathology, NSW Health PathologyRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - Sooin Byeon
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
| | - Angela Chou
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Department of Anatomical Pathology, NSW Health PathologyRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - Sarah Maloney
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
| | - Elizabeth Moon
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
| | - Nick Pavlakis
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Sydney Cancer Center, Royal North Shore HospitalSt LeonardsNew South WalesAustralia
- Northern Cancer InstituteSt LeonardsNew South WalesAustralia
| | - Anthony J. Gill
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Department of Anatomical Pathology, NSW Health PathologyRoyal North Shore HospitalSydneyNew South WalesAustralia
| | - Jaswinder Samra
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
- Upper Gastrointestinal Surgical UnitRoyal North Shore Hospital and North Shore Private HospitalSt LeonardsNew South WalesAustralia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySt LeonardsNew South WalesAustralia
- Northern Clinical School, Kolling Institute of Medical ResearchUniversity of SydneySt LeonardsNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
- Upper Gastrointestinal Surgical UnitRoyal North Shore Hospital and North Shore Private HospitalSt LeonardsNew South WalesAustralia
- The University of Notre Dame AustraliaSydneyNew South WalesAustralia
| |
Collapse
|
34
|
TGF-β2 antisense oligonucleotide enhances T-cell mediated anti-tumor activities by IL-2 via attenuation of fibrotic reaction in a humanized mouse model of pancreatic ductal adenocarcinoma. Biomed Pharmacother 2023; 159:114212. [PMID: 36610224 DOI: 10.1016/j.biopha.2022.114212] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/20/2022] [Accepted: 12/31/2022] [Indexed: 01/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers, with high mortality and recurrence rate. In this study, we generated a human immune system mouse model by transplanting human peripheral blood mononuclear cells into NSG-B2m mice followed by xenografting AsPC-1 cells, after which we assessed the role of transforming growth factor-β2 (TGF-β2) in T-cell-mediated anti-tumor immunity. We observed that inhibiting the TGF-β2 production by TGF-β2 antisense oligonucleotide (TASO) combined with IL-2 delays pancreatic cancer growth. Co-treatment of TASO and IL-2 had little effect on the SMAD-dependent pathway, but significantly inhibited the Akt phosphorylation and sequentially activated GSK-3β. Activation of GSK-3β by TASO subsequently suppressed β-catenin and α-SMA expression and resulted in attenuated fibrotic reactions, facilitating the infiltration of CD8 + cytotoxic T lymphocytes (CTLs) into the tumor. TGF-β2 inhibition suppressed the Foxp3 + regulatory T-cells in peripheral blood and tumors, thereby enhancing the tumoricidal effects of CTLs associated with increased granzyme B and cleaved caspase-3. Moreover, changes in the T-cell composition in peripheral blood and at the tumor site by TASO and IL-2 induced the increase of pro-inflammatory cytokines such as IFN-γ and TNF-α and the decrease of anti-inflammatory cytokines such as TGF-βs. These results indicate that the TGF-β2 inhibition by TASO combined with IL-2 enhances the T-cell mediated anti-tumor immunity against SMAD4-mutated PDAC by modulating the tumor-associated fibrosis, suggesting that TASO in combination with IL-2 may be a promising immunotherapeutic intervention for PDAC.
Collapse
|
35
|
Identification and Validation of Potentially Clinically Relevant CpG Regions within the Class 2 Tumor Suppressor Gene SFRP1 in Pancreatic Cancer. Cancers (Basel) 2023; 15:cancers15030683. [PMID: 36765639 PMCID: PMC9913221 DOI: 10.3390/cancers15030683] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
In pancreatic cancer treatment, tumor stage-dependent chemotherapies are used to prolong overall survival. By measuring DNA promoter hypermethylation in the plasma of patients with stage IV pancreatic cancer, it was recently shown that promoter DNA methylation of the tumor suppressor gene SFRP1 has a high value for predicting failure of drug treatment with gemcitabine. In this study, we therefore aimed to identify as precisely as possible the region in the SFRP1 promoter that is frequently hypermethylated in pancreatic cancer tissue. First, we used the TCGA data set to define CpG-rich regions flanking the SFRP1 transcription start site that were significantly more methylated in pancreatic cancer compared to normal pancreatic acinar tissue. A core CpG island was identified that exhibited abundant tumor DNA methylation and anti-correlation of SFRP1 mRNA expression. To validate our in silico results, we performed bisulfide conversion followed by DNA pyrosequencing of 28 matched formalin-fixed, paraffin-embedded (FFPE) pancreatic cancer cases and six pancreatic cancer cell lines. A defined block of seven CpG sites within the core CpG island was identified, which confirmed our in silico results by showing significantly higher SFRP1 methylation in pancreatic cancer specimens than in normal pancreatic tissue. By selecting this core CpG island, we were able to determine a median overall survival benefit for the low SFRP1 methylation group compared to the high SFRP1 methylation group (702 versus 517 days, p = 0.01) in the TCGA pancreatic cancer cohort. We propose a compact pyrosequencing assay that can be used in the future to further investigate the prognostic value of SFRP1 promoter hypermethylation in predicting pancreatic cancer chemoresistance. Therefore, instead of DNA analysis from blood (liquid biopsy), DNA easily extractable from cancer tissue blocks (FFPE material) could be used.
Collapse
|
36
|
Hu B, Yang Y, Tu J, Cai H, Yang S, Chen X, Chen G. Berbamine Exerts an Anti-oncogenic Effect on Pancreatic Cancer by Regulating Wnt and DNA Damage-related Pathways. Anticancer Agents Med Chem 2023; 23:201-209. [PMID: 35579129 DOI: 10.2174/1871520622666220509174306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 02/08/2023]
Abstract
OBJECTIVE This study aimed to determine the effects of berbamine on pancreatic cancer as well as the underlying mechanisms. METHODS The pancreatic cancer cells were treated with different concentrations of berbamine and then subjected to cell viability assay, colony formation assay, cell cycle analysis, and apoptosis detection. Western blotting and immunofluorescence analyses were performed to investigate the mechanisms underlying the biological effects of berbamine on the pancreatic cancer cells. Furthermore, the in vivo anti-pancreatic cancer effect of berbamine was examined using a mouse xenograft model. RESULTS Berbamine significantly inhibited the proliferation and colony-forming ability of BxPC3 and PANC-1 pancreatic cancer cells while inducing a cell cycle arrest and apoptosis. Moreover, berbamine decreased the expression of β- catenin and phosphorylation of GSK3β but increased the expression of γ-H2AX and 53BP1. Meanwhile, in vivo studies revealed that berbamine attenuated the growth of xenograft tumors derived from PANC-1 cells. Notably, berbamine treatment led to an increase in the expression of Cleaved Caspase 3 and γ-H2AX, as well as a decrease in the expression of Ki-67 and β-catenin in the tumor xenografts. CONCLUSION Berbamine exerts an anti-pancreatic cancer effect, possibly by regulating Wnt and DNA damage-related pathways, suggestive of its therapeutic potential for pancreatic cancer.
Collapse
Affiliation(s)
- Bingren Hu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou 325000, China
| | - Yingnan Yang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou 325000, China
| | - Jinfu Tu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou 325000, China
| | - Huajie Cai
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou 325000, China
| | - Shouzhang Yang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou 325000, China
| | - Xinwei Chen
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou 325000, China
| | - Gang Chen
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou 325000, China
| |
Collapse
|
37
|
Sardar M, Recio-Boiles A, Mody K, Karime C, Chandana SR, Mahadevan D, Starr J, Jones J, Borad M, Babiker H. Pharmacotherapeutic options for pancreatic ductal adenocarcinoma. Expert Opin Pharmacother 2022; 23:2079-2089. [PMID: 36394449 DOI: 10.1080/14656566.2022.2149322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy projected to be the 2nd leading cause of cancer related death in the USA by 2030. This manuscript discusses current and evolving treatment approaches in patients with pancreatic cancer. AREAS COVERED PDAC is classified as: a) resectable, b) borderline resectable, c) unresectable (locally advanced and metastatic). The standard of care for patients who present with resectable pancreatic adenocarcinoma is six months of adjuvant modified (m) FOLFIRINOX, gemcitabine plus capecitabine, or single agent gemcitabine. For many reasons, there has been a paradigm shift to employing neoadjuvant chemotherapy. For resectable and borderline resectable patients, we generally start with systemic therapy and reevaluate resectability with subsequent scans specifically when the tumor is located in the head or body of the pancreas. Combined chemoradiation therapy can be employed in select patients. The standard of care for metastatic PDAC is FOLFIRINOX or gemcitabine and nab-paclitaxel. Germline and somatic genomic profiling should be obtained in all patients. Patients with a germline BRCA mutation can receive upfront gemcitabine and cisplatin. EXPERT OPINION Thorough understanding of molecular pathogenesis in PDAC has opened various therapeutic avenues. We remain optimistic that future treatment modalities such as targeted therapies, cellular therapies and immunotherapy will further improve survival in PDAC.
Collapse
Affiliation(s)
- Muhammad Sardar
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, Az, USA
| | - Alejandro Recio-Boiles
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, Az, USA
| | - Kabir Mody
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| | | | | | - Daruka Mahadevan
- Division of Hematology and Oncology, Department of Medicine, University of Texas, San Antonio, Texas, USA
| | - Jason Starr
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| | - Jeremy Jones
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| | - Mitesh Borad
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Phoenix, AZ, USA
| | - Hani Babiker
- Division of Hematology-Oncology, Department of Medicine, Mayo Clinic Cancer Center, Jacksonville, FL, USA
| |
Collapse
|
38
|
Qin C, Li T, Wang Y, Zhao B, Li Z, Li T, Yang X, Zhao Y, Wang W. CHRNB2 represses pancreatic cancer migration and invasion via inhibiting β-catenin pathway. Cancer Cell Int 2022; 22:340. [DOI: 10.1186/s12935-022-02768-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Pancreatic cancer is one of the most lethal disease with highly fatal and aggressive properties. Lymph node ratio (LNR), the ratio of the number of metastatic lymph nodes to the total number of examined lymph nodes, is an important index to assess lymphatic metastasis and predict prognosis, but the molecular mechanism underlying high LNR was unclear.
Methods
Gene expression and clinical information data of pancreatic cancer were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Patients in TCGA were averagely divided into low and high LNR groups. Then, Weighted Gene Co-expression Network Analysis (WGCNA) was utilized to build co-expression network to explore LNR-related modules and hub genes. GO and KEGG analysis was performed to find key pathways related to lymph node metastasis. Next, GSE101448 and the overall survival data in TCGA was employed to further select significant genes from hub genes. Considering the key role of CHRNB2 in LNR and survival, gene set enrichment analysis (GSEA) was applied to find pathways related to CHRNB2 expression in pancreatic cancer. The contribution of CHRNB2 to migrative and invasive ability of pancreatic cancer cells was confirmed by Transwell assays. We finally explored the role of CHRNB2 in EMT and β-catenin pathway via Western Blot.
Results
High LNR was significantly related to high T stages and poor prognosis. In WGCNA, 14 hub genes (COL5A1, FN1, THBS2, etc.) were positively related to high LNR, 104 hub genes (FFAR1, SCG5, TMEM63C, etc.) were negatively related to high LNR. After taking the intersection with GSE101448, 13 genes (CDK5R2, SYT7, CACNA2D2, etc.) which might prevent lymph node metastasis were further selected. Among them, CHRNB2 showed the strongest relationship with long survival. Moreover, CHRNB2 also negatively related to the T stages and LNR. Next, knockdown of CHRNB2 expression could acetylcholine (ACh)-independently increase the migration and invasion of pancreatic cancer cells, while CHRNB2 overexpression ACh-independently decrease the migration and invasion of pancreatic cancer cells. For exploring the underlying mechanism, CHRNB2 downregulated β-catenin pathway might through controlling its upstream regulators such as SOX6, SRY, SOX17, and TCF7L2.
Conclusions
CHRNB2 negatively relates to lymph node metastasis in pancreatic cancer patients. CHRNB2 could inhibit β-catenin pathway, EMT, migration and invasion of pancreatic cancer cells via ACh-independent mechanism.
Collapse
|
39
|
Chen LJ, Wu L, Wang W, Zhai LL, Xiang F, Li WB, Tang ZG. Long non‑coding RNA 01614 hyperactivates WNT/β‑catenin signaling to promote pancreatic cancer progression by suppressing GSK‑3β. Int J Oncol 2022; 61:116. [PMID: 35929518 PMCID: PMC9387559 DOI: 10.3892/ijo.2022.5406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/24/2022] [Indexed: 11/06/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal type of cancer for which effective therapies are limited. Long non-coding RNAs (lncRNAs) represent a critical type of regulator category, mediating the tumorigenesis and development of various tumor types, including PC. However, the expression patterns and functions of numerous lncRNAs in PC remain poorly understood. In the present study, linc01614 was identified as a PC-related lncRNA. linc01614 was notably upregulated in PC tissues and cell lines and was associated with the poor disease-free survival of patients with PC according to the analysis of The Cancer Genome Atlas-derived datasets. Functionally, linc01614 knockdown suppressed PC cell proliferation, migration and invasion in vitro, and inhibited tumor proliferation in vitro and in vivo. Mechanistically, linc01614 overexpression stabilized the level of β-catenin protein to hyperactivate the WNT/β-catenin signaling pathway in PC cells. Further analyses revealed that linc01614 bound to GSK-3β and perturbed the interaction between GSK-3β and AXIN1, thereby preventing the formation of the β-catenin degradation complex and reducing the degradation of β-catenin. In summary, the present findings reveal that linc01614 may function as an oncogene and promote the progression of PC and may thus be considered as a potential therapeutic target in the future.
Collapse
Affiliation(s)
- Long-Jiang Chen
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lun Wu
- Department of Breast and Τhyroid Surgery, Experiment Center of Medicine, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442008, P.R. China
| | - Wei Wang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lu-Lu Zhai
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Feng Xiang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei-Bo Li
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhi-Gang Tang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
40
|
Islam MS, Morshed MR, Babu G, Khan MA. The role of inflammations and EMT in carcinogenesis. ADVANCES IN CANCER BIOLOGY - METASTASIS 2022; 5:100055. [DOI: 10.1016/j.adcanc.2022.100055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
|
41
|
Development of a Penetratin-Conjugated Stapled Peptide that Inhibits Wnt/β-Catenin Signaling. Bioorg Med Chem 2022; 73:117021. [DOI: 10.1016/j.bmc.2022.117021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022]
|
42
|
Wang P, Deng Z, Li A, Li R, Huang W, Cui J, Chen S, Li B, Zhang S. β-Catenin promotes long-term survival and angiogenesis of peripheral blood mesenchymal stem cells via the Oct4 signaling pathway. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1434-1449. [PMID: 36050404 PMCID: PMC9535028 DOI: 10.1038/s12276-022-00839-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 04/06/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022]
Abstract
Stem cell therapy has been extensively studied to improve heart function following myocardial infarction; however, its therapeutic potency is limited by low rates of engraftment, survival, and differentiation. Here, we aimed to determine the roles of the β-catenin/Oct4 signaling axis in the regulation of long-term survival and angiogenesis of peripheral blood mesenchymal stem cells (PBMSCs). These cells were obtained from rat abdominal aortic blood. We showed that β-catenin promotes the self-renewal, antiapoptotic effects, and long-term survival of PBMSCs by activating the Oct4 pathway through upregulation of the expression of the antiapoptotic factors Bcl2 and survivin and the proangiogenic cytokine bFGF and suppression of the levels of the proapoptotic factors Bax and cleaved caspase-3. β-Catenin overexpression increased Oct4 expression. β-Catenin knockdown suppressed Oct4 expression in PBMSCs. However, β-catenin levels were not affected by Oct4 overexpression or knockdown. Chromatin immunoprecipitation assays proved that β-catenin directly regulates Oct4 transcription in PBMSCs. In vivo, PBMSCs overexpressing β-catenin showed high survival in infarcted hearts and resulted in better myocardial repair. Further functional analysis identified Oct4 as the direct upstream regulator of Ang1, bFGF, HGF, VEGF, Bcl2, and survivin, which cooperatively drive antiapoptosis and angiogenesis of engrafted PBMSCs. These findings revealed the regulation of β-catenin in PBMSCs by the Oct4-mediated antiapoptotic/proangiogenic signaling axis and provide a breakthrough point for improving the long-term survival and therapeutic effects of PBMSCs. Boosting expression of a specific gene has allowed researchers to generate stem cells with increased capacity for tissue repair after a heart attack. Several studies have shown that treatment with a population of circulating cells known as ‘peripheral blood mesenchymal stem cells’ (PBMSCs) can regenerate cardiac tissue. These cells generally have a short lifespan when used therapeutically, but researchers led by Shaoheng Zhang at Jinan University in Guangzhou China have increased long-term survival and performance by boosting expression of the gene encoding β-catenin, a protein that promotes cell survival and proliferation. PBMSCs expressing increased levels of β-catenin preserved heart function in a rat model of heart attack, stimulating blood vessel growth and improving animal survival. This study also reveals proteins regulated by β-catenin, which could potentially be exploited for finer control of PBMSC function.
Collapse
Affiliation(s)
- Pengzhen Wang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China.,Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Zhanyu Deng
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Aiguo Li
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Rongsen Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Weiguang Huang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Jin Cui
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Songsheng Chen
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Biao Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Shaoheng Zhang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China.
| |
Collapse
|
43
|
Nguyen TTP, Suman KH, Nguyen TB, Nguyen HT, Do DN. The Role of miR-29s in Human Cancers—An Update. Biomedicines 2022; 10:biomedicines10092121. [PMID: 36140219 PMCID: PMC9495592 DOI: 10.3390/biomedicines10092121] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that directly bind to the 3’ untranslated region (3’-UTR) of the target mRNAs to inhibit their expression. The miRNA-29s (miR-29s) are suggested to be either tumor suppressors or oncogenic miRNAs that are strongly dysregulated in various types of cancer. Their dysregulation alters the expression of their target genes, thereby exerting influence on different cellular pathways including cell proliferation, apoptosis, migration, and invasion, thereby contributing to carcinogenesis. In the present review, we aimed to provide an overview of the current knowledge on the miR-29s biological network and its functions in cancer, as well as its current and potential applications as a diagnostic and prognostic biomarker and/or a therapeutic target in major types of human cancer.
Collapse
Affiliation(s)
- Thuy T. P. Nguyen
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kamrul Hassan Suman
- Department of Fisheries, Ministry of Fisheries and Livestock, Dhaka 1205, Bangladesh
| | - Thong Ba Nguyen
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Ha Thi Nguyen
- Institute of Research and Development, Duy Tan University, Danang 550000, Vietnam
- Center for Molecular Biology, College of Medicine and Pharmacy, Duy Tan University, Danang 550000, Vietnam
- Correspondence: (H.T.N.); (D.N.D.)
| | - Duy Ngoc Do
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS B2N 5E3, Canada
- Correspondence: (H.T.N.); (D.N.D.)
| |
Collapse
|
44
|
Ye D, Liu Y, Chen Y, Li G, Sun B, Peng J, Xu Q. Identification of lncRNA biomarkers in hepatocellular carcinoma by comprehensive analysis of the lncRNA-mediated ceRNA network. Front Genet 2022; 13:832952. [PMID: 36105104 PMCID: PMC9465287 DOI: 10.3389/fgene.2022.832952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022] Open
Abstract
Growing evidence implicates that miRNAs can interact with long non-coding RNAs (lncRNAs) to regulate target mRNAs through competitive interactions. However, this mechanism that regulate tumorigenesis and cancer progression remains largely unexplored. Long non-coding RNAs (lncRNAs) act as competing endogenous RNAs (ceRNAs), which play a significant role in regulating gene expression. The purpose of our study was to determine potential lncRNA biomarkers to predict the prognosis of HCC by comprehensive analysis of a ceRNA network. The edgeR package was used to obtain the differentially expressed RNA datasets by analyzing 370 HCC tissues and 50 adjacent non-HCC tissues from The Cancer Genome Atlas (TCGA). Through investigating the differentially expressed between HCC tissues and adjacent non-HCC tissues, a total of 947 lncRNAs, 52 miRNAs, and 1,650 mRNAs were obtained. The novel constructed ceRNA network incorporated 99 HCC-specific lncRNAs, four miRNAs, and 55 mRNAs. Survival analysis identified 22 differentially expressed mRNAs, four miRNAs, and nine lncRNAs which were associated with overall survival (OS) time in HCC (p < 0.05), and further exploration was performed to assess the correlation of these differentially expressed genes with tumor stage. The Interpretation of the potential functions of these differentially expressed genes in HCC was realized by Gene ontology (GO) and KEGG pathway enrichment analyses. Seven lncRNAs were confirmed based on univariate Cox regression analysis, lasso COX regression analysis and multivariate Cox regression analysis to construct a predictive model in HCC patients which were related to the prognosis of OS. In summary, ceRNAs contributed to explore the mechanism of tumorigenesis and development, and a model with seven lncRNAs might be potential biomarker to predict the prognosis of HCC. These findings supported the need to studies on the mechanisms involved in the regulation of HCC by ceRNAs.
Collapse
Affiliation(s)
- Dingde Ye
- Medicine School of Southeast University Nanjing Drum Tower Hospital, Nanjing, China
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yaping Liu
- School of Life Science and Technology, Southeast University, Nanjing, China
| | - Yanuo Chen
- Medicine School of Southeast University Nanjing Drum Tower Hospital, Nanjing, China
| | - Guoqiang Li
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Beicheng Sun
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Beicheng Sun, ; Jin Peng, ; Qingxiang Xu,
| | - Jin Peng
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Beicheng Sun, ; Jin Peng, ; Qingxiang Xu,
| | - Qingxiang Xu
- Medicine School of Southeast University Nanjing Drum Tower Hospital, Nanjing, China
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Beicheng Sun, ; Jin Peng, ; Qingxiang Xu,
| |
Collapse
|
45
|
ALCAM: A Novel Surface Marker on EpCAMlow Circulating Tumor Cells. Biomedicines 2022; 10:biomedicines10081983. [PMID: 36009530 PMCID: PMC9405826 DOI: 10.3390/biomedicines10081983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Current strategies in circulating tumor cell (CTC) isolation in pancreatic cancer heavily rely on the EpCAM and cytokeratin cell status. EpCAM is generally not considered a good marker given its transitory change during Epithelial to Mesenchymal Transition (EMT) or reverse EMT. There is a need to identify other surface markers to capture the complete repertoire of PDAC CTCs. The primary objective of the study is to characterize alternate surface biomarkers to EpCAM on CTCs that express low or negligible levels of surface EpCAM in pancreatic cancer patients. Methods: Flow cytometry and surface mass spectrometry were used to identify proteins expressed on the surface of PDAC CTCs in culture. CTCs were grown under conditions of attachment and in co-culture with naïve neutrophils. Putative biomarkers were then validated in GEMMs and patient samples. Results: Surface proteomic profiling of CTCs identified several novel protein biomarkers. ALCAM was identified as a novel robust marker in GEMM models and in patient samples. Conclusions: We identified several novel surface biomarkers on CTCs expressed under differing conditions of culture. ALCAM was validated and identified as a novel alternate surface marker on EpCAMlow CTCs.
Collapse
|
46
|
Jiang J, Cheng Y, Dai S, Zou B, Guo X. Suppression of rhomboid domain-containing 1 produces anticancer effects in pancreatic adenocarcinoma through affection of the AKT/GSK-3β/β-catenin pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:1944-1956. [PMID: 35442567 DOI: 10.1002/tox.23541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/20/2022] [Accepted: 04/10/2022] [Indexed: 06/14/2023]
Abstract
The protumor role of rhomboid domain-containing 1 (RHBDD1) has been observed in multiple cancers. However, the relationship between RHBDD1 and pancreatic adenocarcinoma has not been addressed. This project focused on the potential relevance of RHBDD1 in pancreatic adenocarcinoma. Bioinformatic analysis by publicly available data revealed that RHBDD1 was abundantly expressed in pancreatic adenocarcinoma. We further verified that RHBDD1 was expressed highly in clinical specimens of pancreatic adenocarcinoma. The Kaplan-Meier curve demonstrated that high-RHBDD1 expression was associated with poor prognosis in pancreatic adenocarcinoma patients. The functional studies revealed that depletion of RHBDD1 produced in vitro anticancer effects in pancreatic adenocarcinoma cells, including retardation of proliferation, reduction of metastatic potential, and induction of cell-cycle arrest at the G0/G1 phase and apoptosis. Mechanistic studies indicated that loss of RHBDD1 affected the activation of β-catenin via regulation of AKT. Forced expression of β-catenin reversed the RHBDD1-loss-induced anticancer effects in pancreatic adenocarcinoma cells. Crucially, depletion of RHBDD1 retarded the growth of pancreatic adenocarcinoma xenografts in vivo, a phenomenon associated with the AKT/β-catenin pathway. Collectively, these findings delineated that restraint of RHBDD1 displayed remarkable anticancer effects in pancreatic adenocarcinoma by affecting the AKT/β-catenin pathway. Our work unveils a pivotal role of RHBDD1 in pancreatic adenocarcinoma and proposes it as a novel candidate target for anticancer therapy of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Jiong Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Cheng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shejiao Dai
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Baicang Zou
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
47
|
Ma Y, Di Y, Li Q, Zhan Q, He X, Liu S, Zou H, Corpe C, Chen L, Wang J. LncRNAs as epigenetic regulators of epithelial to mesenchymal transition in pancreatic cancer. Discov Oncol 2022; 13:61. [PMID: 35819532 PMCID: PMC9276894 DOI: 10.1007/s12672-022-00522-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/01/2022] [Indexed: 11/04/2022] Open
Abstract
Pancreatic cancer is the leading cause of cancer-related mortality because of tumor metastasis. Activation of the epithelial-to-mesenchymal transition (EMT) pathway has been confirmed to be an important driver of pancreatic cancer progression from initiation to metastasis. Long noncoding RNAs (lncRNAs) have been reported to exert essential physiological functions in pancreatic cancer progression by regulating the EMT program. In this review, we have summarized the role of EMT-related lncRNAs in human pancreatic cancer and the potential molecular mechanisms by which lncRNAs can be vital epigenetic regulators of epithelial to mesenchymal transition. Specifically, EMT-activating transcription factors (EMT-TFs) regulate EMT via TGF-β/Smad, Wnt/β-catenin, and JAK/STAT pathways. In addition, the interaction between lncRNAs and HIF-1α and m6A RNA methylation also have an impact on tumor metastasis and EMT in pancreatic cancer. This review will provide insights into lncRNAs as promising biomarkers for tumor metastasis and potential therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Yan Ma
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Yang Di
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiuyue Li
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Qilin Zhan
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Xiaomeng He
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Shanshan Liu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Heng Zou
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Christopher Corpe
- King's College London, Nutritional Science Department, 150 Stamford Street, Waterloo, London, SE19NH, UK
| | - Litian Chen
- Department of Hepatobiliary Surgery, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Kongjiang Road 1665, Shanghai, China.
| | - Jin Wang
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, People's Republic of China.
| |
Collapse
|
48
|
Effects of TP53 Mutations and miRs on Immune Responses in the Tumor Microenvironment Important in Pancreatic Cancer Progression. Cells 2022; 11:cells11142155. [PMID: 35883598 PMCID: PMC9318640 DOI: 10.3390/cells11142155] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 01/27/2023] Open
Abstract
Approximately 90% of pancreatic cancers are pancreatic ductal adenocarcinomas (PDAC). PDAC is the fourth leading cause of cancer death world-wide. Therapies for PDAC are largely ineffective due to the dense desmoplastic tumor microenvironment which prevents chemotherapeutic drugs and small molecule inhibitors from exerting effective anti-cancer effects. In this review, we will discuss the roles of TP53 and miRs on the PDAC tumor microenvironment and how loss of the normal functions of TP53 promote tumor progression. The TP53 gene is mutated in approximately 50% of pancreatic cancers. Often, these TP53 mutations are point mutations which confer additional functions for the TP53 proteins. These are called gain of function (GOF) mutations (mut). Another class of TP53 mutations are deletions which result in loss of the TP53 protein; these are referred to TP53-null mutations. We have organized this review into various components/properties of the PDAC microenvironment and how they may be altered in the presence of mutant TP53 and loss of certain miR expression.
Collapse
|
49
|
Mehra S, Singh S, Nagathihalli N. Emerging Role of CREB in Epithelial to Mesenchymal Plasticity of Pancreatic Cancer. Front Oncol 2022; 12:925687. [PMID: 35800049 PMCID: PMC9253527 DOI: 10.3389/fonc.2022.925687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive solid malignancy with a high rate of metastasis and therapeutic resistance as its major hallmarks. Although a defining mutational event in pancreatic cancer initiation is the presence of oncogenic KRAS, more advanced PDAC lesions accumulate additional genomic alterations, including loss of tumor suppressor gene TP53. Co-occurrence of mutant KRAS and TP53 in PDAC promotes hyperactivation of cancer cell signaling pathways driving epithelial to mesenchymal plasticity (EMP). The cellular process of EMP influences the biological behavior of cancer cells by increasing their migratory and invasive properties, thus promoting metastasis. Our previous work has demonstrated that oncogenic KRAS-mediated activation of cyclic AMP response element-binding protein 1 (CREB) is one of the critical drivers of PDAC aggressiveness. The therapeutic approach of targeting this key transcription factor attenuates tumor burden in genetically engineered mouse models (GEMMs) of this disease. Herein, we discuss the significant role of CREB in perpetuating disease aggressiveness and therapeutic resistance through the EMP process. Furthermore, this review updates the therapeutic implications of targeting CREB, highlighting the challenges and emerging approaches in PDAC.
Collapse
Affiliation(s)
- Siddharth Mehra
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Samara Singh
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nagaraj Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
- *Correspondence: Nagaraj Nagathihalli,
| |
Collapse
|
50
|
Nisar M, Paracha RZ, Adil S, Qureshi SN, Janjua HA. An Extensive Review on Preclinical and Clinical Trials of Oncolytic Viruses Therapy for Pancreatic Cancer. Front Oncol 2022; 12:875188. [PMID: 35686109 PMCID: PMC9171400 DOI: 10.3389/fonc.2022.875188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy resistance and peculiar tumor microenvironment, which diminish or mitigate the effects of therapies, make pancreatic cancer one of the deadliest malignancies to manage and treat. Advanced immunotherapies are under consideration intending to ameliorate the overall patient survival rate in pancreatic cancer. Oncolytic viruses therapy is a new type of immunotherapy in which a virus after infecting and lysis the cancer cell induces/activates patients’ immune response by releasing tumor antigen in the blood. The current review covers the pathways and molecular ablation that take place in pancreatic cancer cells. It also unfolds the extensive preclinical and clinical trial studies of oncolytic viruses performed and/or undergoing to design an efficacious therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Maryum Nisar
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Sidra Adil
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | | | - Hussnain Ahmed Janjua
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, Pakistan
| |
Collapse
|