1
|
Kamble OS, Chatterjee R, Abishek KG, Chandra J, Alsayari A, Wahab S, Sahebkar A, Kesharwani P, Dandela R. Small molecules targeting mitochondria as an innovative approach to cancer therapy. Cell Signal 2024; 124:111396. [PMID: 39251050 DOI: 10.1016/j.cellsig.2024.111396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cellular death evasion is a defining characteristic of human malignancies and a significant contributor to therapeutic inefficacy. As a result of oncogenic inhibition of cell death mechanisms, established therapeutic regimens seems to be ineffective. Mitochondria serve as the cellular powerhouses, but they also function as repositories of self-destructive weaponry. Changes in the structure and activities of mitochondria have been consistently documented in cancer cells. In recent years, there has been an increasing focus on using mitochondria as a targeted approach for treating cancer. Considerable attention has been devoted to the development of delivery systems that selectively aim to deliver small molecules called "mitocans" to mitochondria, with the ultimate goal of modulating the physiology of cancer cells. This review summarizes the rationale and mechanism of mitochondrial targeting with small molecules in the treatment of cancer, and their impact on the mitochondria. This paper provides a concise overview of the reasoning and mechanism behind directing treatment towards mitochondria in cancer therapy, with a particular focus on targeting using small molecules. This review also examines diverse small molecule types within each category as potential therapeutic agents for cancer.
Collapse
Affiliation(s)
- Omkar S Kamble
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Rana Chatterjee
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - K G Abishek
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| |
Collapse
|
2
|
Luo M, Wang YM, Zhao FK, Luo Y. Recent Advances in Nanomaterial-Mediated Cell Death for Cancer Therapy. Adv Healthc Mater 2024:e2402697. [PMID: 39498722 DOI: 10.1002/adhm.202402697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/01/2024] [Indexed: 11/07/2024]
Abstract
Nanomedicine has shown great anticancer potential by disrupting redox homeostasis and increasing the levels of oxidative stress, but the therapeutic effect is limited by factors including the intrinsic self-protection mechanism of tumors. Cancer cell death can be induced by the exploration of different cell death mechanisms, such as apoptosis, pyroptosis, necroptosis, cuproptosis, and ferroptosis. The merging of nanotechnology with biomedicine has provided tremendous opportunities to construct cell death-based nanomedicine for innovative cancer therapy. Nanocarriers are not only used for the targeted delivery of cell death inducers, but also as therapeutic components to induce cell death to achieve efficient tumor treatment. This review focuses on seven cell death modalities mediated by nanomaterials, such as apoptosis, pyroptosis, necroptosis, ferroptosis, cuprotosis, immunogenic cell death, and autophagy. The mechanisms of these seven cell death modalities are described in detail, as well as the preparation of nanomaterials that induce them and the mechanisms, they used to exert their effects. Finally, this work describes the potential future development based on the current knowledge related to cell death induced by nanomaterials.
Collapse
Affiliation(s)
- Min Luo
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yuan-Min Wang
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Fu-Kun Zhao
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yong Luo
- Department of Neurology, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| |
Collapse
|
3
|
Moon J, Kim SW, Lee JS. Metal Ion-Condensed DNA Nanoparticle Library: Phase Separation and Transition and Antisense Therapy Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59116-59127. [PMID: 39427257 DOI: 10.1021/acsami.4c16869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
DNA condensation has long been investigated as a fundamental cellular activity and is known to be driven by the mediation of diverse condensing agents. The phase behaviors of DNA during condensation are particularly interesting because the complicated molecular structure of natural nucleotides fundamentally allows electrostatic, coordinate covalent, and various other secondary interactions with the condensing agents. Recently, metal ion (Mn+)-induced DNA condensation has emerged as a powerful approach to synthesizing nanoparticulate DNA structures suitable for therapeutic gene delivery. However, how the DNA phase changes during Mn+-induced DNA condensation has rarely been observed and is not understood yet. In this study, a library of Mn+-condensed DNA nanoparticles (Mn+-CDNPs) was established using 30 different types of Mn+s, and their phase behaviors during condensation were elucidated using spherical nucleic acids (SNAs) as electron microscopic labels. Importantly, the phase transition and separation of DNA were demonstrated to be driven by the Mn+s into either the growth of individual DNA particles or the fission of bulky DNA aggregates. Pt2+ and Eu3+ were chosen as model systems for the demonstration. The hard and soft acid nature of Mn+ is presumably the underlying driving force of these phase transitions. In addition, the Mn+-controlled anticancer therapeutic efficiency of the Mn+-CDNP library as a state-of-the-art gene delivery platform was demonstrated even for unmodified antisense oligonucleotides in association with the potential toxicity of the Mn+s released from the Mn+-CDNPs. This comprehensive study of the Mn+-dependent condensation of nucleic acids provides profound insights into the chemistry of the nucleic acid-Mn+ interactions and the reliable theragnostic applications of Mn+-CDNPs as functional nucleic acid nanostructures.
Collapse
Affiliation(s)
- Jeesu Moon
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Sang-Won Kim
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jae-Seung Lee
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
4
|
Galla MS, Kale NB, Sharma A, Hajare A, Godugu C, Shankaraiah N. Development of chromone-thiazolidine-2,4-dione Knoevenagel conjugates as apoptosis inducing agents. Bioorg Med Chem Lett 2024; 109:129853. [PMID: 38909705 DOI: 10.1016/j.bmcl.2024.129853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/28/2024] [Accepted: 06/19/2024] [Indexed: 06/25/2024]
Abstract
Overexpression of Bcl-2 protein is a predominant hallmark of disturbed apoptotic pathway in most of the cancers. Herein, chromone-linked thiazolidinediones were designed and synthesized to target Bcl-2 for regulating anti-apoptotic proteins. The study on in vitro cancer cell lines revealed the presence of compounds 8a, 8k, 8l, and 8n, which were found to have good to moderate anti-proliferative activity (with an IC50 concentration less than 10 µM). Among them, 8l depicted the highest cytotoxicity on the A549 cell line with an IC50 of 6.1 ± 0.02 µM. Aberrantly, the compounds displayed less toxicity towards human embryonic kidney HEK cells underlining its selectivity. The DCFDA study revealed a gradual increase in the ROS generation of 8l, followed by its quantification by flow analysis. Similarly, the studies including DAPI, AO/EtBr and Annexin-V binding clearly elucidated the DNA damage, membrane integrity prospects, and insights for early and late apoptotic phases. Markedly, the Bcl-2-FITC anti-body study revealed that compound 8l reduced the expression of anti-apoptotic proteins by 79.1 % compared to the control at 9 µM concentration. In addition, the molecular docking study provided the impending scope of these hybrids, showing promising interaction with the Mcl-1 target (member of the Bcl-2 family) with comparable binding affinities.
Collapse
Affiliation(s)
- Mary Sravani Galla
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Nandini B Kale
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Aditya Hajare
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
5
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
6
|
Çakan E, Lara OD, Szymanowska A, Bayraktar E, Chavez-Reyes A, Lopez-Berestein G, Amero P, Rodriguez-Aguayo C. Therapeutic Antisense Oligonucleotides in Oncology: From Bench to Bedside. Cancers (Basel) 2024; 16:2940. [PMID: 39272802 PMCID: PMC11394571 DOI: 10.3390/cancers16172940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/15/2024] Open
Abstract
Advancements in our comprehension of tumor biology and chemoresistance have spurred the development of treatments that precisely target specific molecules within the body. Despite the expanding landscape of therapeutic options, there persists a demand for innovative approaches to address unmet clinical needs. RNA therapeutics have emerged as a promising frontier in this realm, offering novel avenues for intervention such as RNA interference and the utilization of antisense oligonucleotides (ASOs). ASOs represent a versatile class of therapeutics capable of selectively targeting messenger RNAs (mRNAs) and silencing disease-associated proteins, thereby disrupting pathogenic processes at the molecular level. Recent advancements in chemical modification and carrier molecule design have significantly enhanced the stability, biodistribution, and intracellular uptake of ASOs, thereby bolstering their therapeutic potential. While ASO therapy holds promise across various disease domains, including oncology, coronary angioplasty, neurological disorders, viral, and parasitic diseases, our review manuscript focuses specifically on the application of ASOs in targeted cancer therapies. Through a comprehensive examination of the latest research findings and clinical developments, we delve into the intricacies of ASO-based approaches to cancer treatment, shedding light on their mechanisms of action, therapeutic efficacy, and prospects.
Collapse
Affiliation(s)
- Elif Çakan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Olivia D Lara
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Emine Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Medical Biology, Faculty of Medicine, University of Gaziantep, Gaziantep 27310, Turkey
| | | | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
7
|
Kong Q, Zhu Q, Yang Y, Wang W, Qian J, Chen Y. Current status and trend of mitochondrial research in lung cancer: A bibliometric and visualization analysis. Heliyon 2024; 10:e34442. [PMID: 39144972 PMCID: PMC11320136 DOI: 10.1016/j.heliyon.2024.e34442] [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: 05/09/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
This study summarizes and analyzes the relationship between mitochondria and the pathogenesis of lung cancer. The related articles in the Web of Science core literature database are searched and collected, and the data are processed by R software, Citespace, VOSviewer, and Excel. A total of 4476 related papers were retrieved, 4476 articles from 20162 co-authors of 3968 institutions in 84 countries and published in 951 journals. Through various bibliometric analysis tools, the relationship between mitochondria and the pathogenesis of lung cancer was analyzed, the previous research results were summarized, and the potential research direction was found.
Collapse
Affiliation(s)
- Qing Kong
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Qingyong Zhu
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Yuxia Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Wei Wang
- Clinical Medical College, Weifang Medical University, Weifang, 261053, PR China
| | - Juan Qian
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| | - Yong Chen
- Functional Examination Department, Northern Jiangsu People's Hospital, Affiliated to Yangzhou University, Yangzhou, 225001, PR China
| |
Collapse
|
8
|
Chen P, Ding N, Pan D, Chen X, Li S, Luo Y, Chen Z, Xu Y, Zhu X, Wang K, Zou W. PET imaging for the early evaluation of ocular inflammation in diabetic rats by using [ 18F]-DPA-714. Exp Eye Res 2024; 245:109986. [PMID: 38945519 DOI: 10.1016/j.exer.2024.109986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Ocular complications of diabetes mellitus (DM) are the leading cause of vision loss. Ocular inflammation often occurs in the early stage of DM; however, there are no proven quantitative methods to evaluate the inflammatory status of eyes in DM. The 18 kDa translocator protein (TSPO) is an evolutionarily conserved cholesterol binding protein localized in the outer mitochondrial membrane. It is a biomarker of activated microglia/macrophages; however, its role in ocular inflammation is unclear. In this study, fluorine-18-DPA-714 ([18F]-DPA-714) was evaluated as a specific TSPO probe by cell uptake, cell binding assays and micro positron emission tomography (microPET) imaging in both in vitro and in vivo models. Primary microglia/macrophages (PMs) extracted from the cornea, retina, choroid or sclera of neonatal rats with or without high glucose (50 mM) treatment were used as the in vitro model. Sprague-Dawley (SD) rats that received an intraperitoneal administration of streptozotocin (STZ, 60 mg/kg once) were used as the in vivo model. Increased cell uptake and high binding affinity of [18F]-DPA-714 were observed in primary PMs under hyperglycemic stress. These findings were consistent with cellular morphological changes, cell activation, and TSPO up-regulation. [18F]-DPA-714 PET imaging and biodistribution in the eyes of DM rats revealed that inflammation initiates in microglia/macrophages in the early stages (3 weeks and 6 weeks), corresponding with up-regulated TSPO levels. Thus, [18F]-DPA-714 microPET imaging may be an effective approach for the early evaluation of ocular inflammation in DM.
Collapse
Affiliation(s)
- Peng Chen
- Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Jintan Affiliated Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Nannan Ding
- Department of Ophthalmology, Wuxi No.2 People's Hospital, Jiangnan University Medical Center (JUMC), Wuxi, Jiangsu, China; Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China
| | - Donghui Pan
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuelian Chen
- Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, PuNan Branch of Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - ShiYi Li
- Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Jingjiang People's Hospital Affiliated to Yangzhou University, Taizhou, Jiangsu, China
| | - Yidan Luo
- Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China
| | - Ziqing Chen
- Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yuping Xu
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xue Zhu
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ke Wang
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Wenjun Zou
- Department of Ophthalmology, Wuxi No.2 People's Hospital, Jiangnan University Medical Center (JUMC), Wuxi, Jiangsu, China; Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China.
| |
Collapse
|
9
|
Nasimi Shad A, Akhlaghipour I, Alshakarchi HI, Saburi E, Moghbeli M. Role of microRNA-363 during tumor progression and invasion. J Physiol Biochem 2024; 80:481-499. [PMID: 38691273 DOI: 10.1007/s13105-024-01022-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/05/2024] [Indexed: 05/03/2024]
Abstract
Recent progresses in diagnostic and therapeutic methods have significantly improved prognosis in cancer patients. However, cancer is still considered as one of the main causes of human deaths in the world. Late diagnosis in advanced tumor stages can reduce the effectiveness of treatment methods and increase mortality rate of cancer patients. Therefore, investigating the molecular mechanisms of tumor progression can help to introduce the early diagnostic markers in these patients. MicroRNA (miRNAs) has an important role in regulation of pathophysiological cellular processes. Due to their high stability in body fluids, they are always used as the non-invasive markers in cancer patients. Since, miR-363 deregulation has been reported in a wide range of cancers, we discussed the role of miR-363 during tumor progression and metastasis. It has been reported that miR-363 has mainly a tumor suppressor function through the regulation of transcription factors, apoptosis, cell cycle, and structural proteins. MiR-363 also affected the tumor progression via regulation of various signaling pathways such as WNT, MAPK, TGF-β, NOTCH, and PI3K/AKT. Therefore, miR-363 can be introduced as a probable therapeutic target as well as a non-invasive diagnostic marker in cancer patients.
Collapse
Affiliation(s)
- Arya Nasimi Shad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hawraa Ibrahim Alshakarchi
- Al-Zahra Center for Medical and Pharmaceutical Research Sciences (ZCMRS), Al-Zahraa University for Women, Karbala, Iraq
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Lee D, Lee SY, Ra MJ, Jung SM, Yu JN, Kang KS, Kim KH. Cancer therapeutic potential of hovetrichoside C from Jatropha podagrica on apoptosis of MDA-MB-231 human breast cancer cells. Food Chem Toxicol 2024; 190:114794. [PMID: 38849046 DOI: 10.1016/j.fct.2024.114794] [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: 01/04/2024] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Phytochemical analysis of the methanolic extracts of Jatropha podagrica stalks and roots using liquid chromatography-mass spectrometry (LC-MS) led to the isolation of six compounds: corchoionoside C (1), isobiflorin (2), fraxin (3), hovetrichoside C (4), fraxetin (5), and corillagin (6). The isolated compounds (1-6) were tested for their cytotoxicity against MDA-MB-231 human breast cancer cells. Remarkably, compound 4 (hovetrichoside C) exhibited robust cytotoxicity against MDA-MB-231 cells, displaying an IC50 value of 50.26 ± 1.22 μM, along with an apoptotic cell death rate of 24.21 ± 2.08% at 100 μM. Treatment involving compound 4 amplified protein levels of cleaved caspase-8, -9, -3, -7, BH3-interacting domain death agonist (Bid), Bcl-2-associated X protein (Bax), and cleaved poly (ADP-ribose) polymerase (cleaved PARP), while concurrently reducing B-cell lymphoma 2 (Bcl-2) levels. In totality, these findings underscore that hovetrichoside C (4) possesses anti-breast cancer activity that revolves around apoptosis induction via both extrinsic and intrinsic signaling pathways.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam, 13120, Republic of Korea
| | - Seo Yoon Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Moon-Jin Ra
- Hongcheon Institute of Medicinal Herb, Hongcheon-gun, Gangwon-do, 25142, Republic of Korea
| | - Sang-Mi Jung
- Hongcheon Institute of Medicinal Herb, Hongcheon-gun, Gangwon-do, 25142, Republic of Korea
| | - Jeong-Nam Yu
- Nakdonggang National Institute of Biological Resources, Sangju, Gyeongsangbuk-do, 37242, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, 13120, Republic of Korea.
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
11
|
Liu W, Khalid M, Wahab S, Faizan Siddiqui M, Hasan Khan S, Sadiq M, Khatoon Z. A multitier virtual screening study of phytoconstituents as Myeloid Cell Leukemias 1 inhibitors. J Biomol Struct Dyn 2024; 42:5219-5228. [PMID: 37418235 DOI: 10.1080/07391102.2023.2226739] [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: 04/11/2023] [Accepted: 06/09/2023] [Indexed: 07/08/2023]
Abstract
Myeloid Cell Leukemia 1 (MCL1) is an anti-apoptotic protein that plays a critical role in regulating cell survival, particularly in cancer cells. It is a member of the BCL-2 family of proteins, which control the intrinsic pathway of apoptosis. MCL1 has emerged as a promising target for cancer therapy because it is overexpressed in a wide range of cancers, including breast, lung, prostate, and hematologic malignancies. Due to its remarkable role in cancer progression, it has been reflected as a promising drug target for cancer therapy. A few MCL1 inhibitors have been identified previously, but further research is needed to develop novel, effective and safe MCL1 inhibitors that can overcome resistance mechanisms and minimize toxicity in normal cells. In this study, we aim to search for compounds that target the critical binding site of MCL1 from phytoconstituent library from the IMPPAT database. To accomplish this, a multitier virtual screening approach involving molecular docking and molecular dynamics simulations (MDS) were used to evaluate their suitability for the receptor. Notably, certain screened phytoconstituents have appreciable docking scores and stable interactions toward the binding pocket of MCL1. The screened compounds underwent ADMET and bioactivity analysis to establish their anticancer properties. One phytoconstituent, Isopongaflavone, was identified that exhibiting higher docking and drug-likeness than the already reported MCL1 inhibitor, Tapotoclax. Isopongaflavone and and Tapotoclax, along with MCL1, were subjected to 100 nanoseconds (ns) MDS study to verify their stability inside the binding site of MCL1. The MDS findings demonstrated a strong binding affinity between Isopongaflavone and the MCL1 binding pocket, resulting in reduced conformational fluctuations. This investigation proposes Isopongaflavone as a promising candidate for the development of innovative anticancer therapeutics, pending the necessary validation procedures. Also, the findings provide valuable information for designing MCL1 inhibitors based on the protein's structure.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wenjun Liu
- School of Environment and Resources, Chongqing Technology and Business University, Chongqing, China
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | | | - Shaheer Hasan Khan
- Enzymology and nanotechnology laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Mohd Sadiq
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Zeenat Khatoon
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
12
|
Thapa D, Kumar V, Naik B, Kumar V, Gupta AK, Mohanta YK, Mishra B, Rustagi S. Harnessing probiotic foods: managing cancer through gut health. Food Sci Biotechnol 2024; 33:2141-2160. [PMID: 39130664 PMCID: PMC11315834 DOI: 10.1007/s10068-024-01638-5] [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: 04/26/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 08/13/2024] Open
Abstract
One of the greatest threats to global health is cancer. Probiotic foods have been shown to have therapeutic promise in the management of cancer, even though traditional treatments such as radiation therapy, chemotherapy, and surgery are still essential. The generation of anticarcinogenic compounds, immune system stimulation, and gut microbiota regulation are a few ways that probiotics when taken in sufficient quantities, might help health. The purpose of this review is to examine the therapeutic potential of probiotic foods in the management of cancer. Research suggests that certain strains of probiotics have anticancer effects by preventing the growth of cancer cells, triggering apoptosis, and reducing angiogenesis in new tumors. Probiotics have shown promise in mitigating treatment-related adverse effects, such as diarrhea, mucositis, and immunosuppression caused by chemotherapy, improving the general quality of life for cancer patients. However, there are several factors, such as patient-specific features, cancer subtype, and probiotic strain type and dosage, which affect how effective probiotic therapies are in managing cancer. More research is necessary to find the long-term safety and efficacy characteristics of probiotics as well as to clarify the best ways to incorporate them into current cancer treatment methods. Graphical abstract Graphical representation showing the role of probiotic foods in cancer management.
Collapse
Affiliation(s)
- Devika Thapa
- Department of Food Science and Technology, Graphic Era Deemed to be University, Clement Town, Dehradun, Uttarakhand 248002 India
| | - Vijay Kumar
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, Uttarakhand 248140 India
| | - Bindu Naik
- Department of Food Science and Technology, Graphic Era Deemed to be University, Clement Town, Dehradun, Uttarakhand 248002 India
- School of Agriculture, Graphic Era Hill University, Dehradun, Uttarakhand India
| | - Vivek Kumar
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, Uttarakhand 248140 India
| | - Arun Kumar Gupta
- Department of Food Science and Technology, Graphic Era Deemed to be University, Clement Town, Dehradun, Uttarakhand 248002 India
| | - Yugal Kishore Mohanta
- Nano-biotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya, Techno City, 9th Mile, Baridua, Ri-Bhoi, Meghalaya 793101 India
| | - Bishwambhar Mishra
- Department of Biotechnology, Chaitanya Bharathi Institute of Technology (CBIT), Gandipet, Hyderabad, Telangana 500075 India
| | - Sarvesh Rustagi
- Department of Food Technology, SALS, Uttaranchal University, Dehradun, 248007 Uttarakhand India
| |
Collapse
|
13
|
Shin HE, Han JH, Shin S, Bae GH, Son B, Kim TH, Park HH, Park CG, Park W. M1-polarized macrophage-derived cellular nanovesicle-coated lipid nanoparticles for enhanced cancer treatment through hybridization of gene therapy and cancer immunotherapy. Acta Pharm Sin B 2024; 14:3169-3183. [PMID: 39027257 PMCID: PMC11252390 DOI: 10.1016/j.apsb.2024.03.004] [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: 01/05/2024] [Revised: 01/25/2024] [Accepted: 02/07/2024] [Indexed: 07/20/2024] Open
Abstract
Optimum genetic delivery for modulating target genes to diseased tissue is a major obstacle for profitable gene therapy. Lipid nanoparticles (LNPs), considered a prospective vehicle for nucleic acid delivery, have demonstrated efficacy in human use during the COVID-19 pandemic. This study introduces a novel biomaterial-based platform, M1-polarized macrophage-derived cellular nanovesicle-coated LNPs (M1-C-LNPs), specifically engineered for a combined gene-immunotherapy approach against solid tumor. The dual-function system of M1-C-LNPs encapsulates Bcl2-targeting siRNA within LNPs and immune-modulating cytokines within M1 macrophage-derived cellular nanovesicles (M1-NVs), effectively facilitating apoptosis in cancer cells without impacting T and NK cells, which activate the intratumoral immune response to promote granule-mediating killing for solid tumor eradication. Enhanced retention within tumor was observed upon intratumoral administration of M1-C-LNPs, owing to the presence of adhesion molecules on M1-NVs, thereby contributing to superior tumor growth inhibition. These findings represent a promising strategy for the development of targeted and effective nanoparticle-based cancer genetic-immunotherapy, with significant implications for advancing biomaterial use in cancer therapeutics.
Collapse
Affiliation(s)
- Ha Eun Shin
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Jun-Hyeok Han
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Deparment of Inteligent Precision Healthcare Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seungyong Shin
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Ga-Hyun Bae
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth, SKKU Institute for Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
| | - Boram Son
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Tae-Hyung Kim
- Department of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Chun Gwon Park
- Deparment of Inteligent Precision Healthcare Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Biomedical Engineering, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth, SKKU Institute for Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
14
|
Wang ZH, Wang J, Liu F, Sun S, Zheng Q, Hu X, Yin Z, Xie C, Wang H, Wang T, Zhang S, Wang YP. THAP3 recruits SMYD3 to OXPHOS genes and epigenetically promotes mitochondrial respiration in hepatocellular carcinoma. FEBS Lett 2024; 598:1513-1531. [PMID: 38664231 DOI: 10.1002/1873-3468.14889] [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: 01/03/2024] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 06/27/2024]
Abstract
Mitochondria harbor the oxidative phosphorylation (OXPHOS) system to sustain cellular respiration. However, the transcriptional regulation of OXPHOS remains largely unexplored. Through the cancer genome atlas (TCGA) transcriptome analysis, transcription factor THAP domain-containing 3 (THAP3) was found to be strongly associated with OXPHOS gene expression. Mechanistically, THAP3 recruited the histone methyltransferase SET and MYND domain-containing protein 3 (SMYD3) to upregulate H3K4me3 and promote OXPHOS gene expression. The levels of THAP3 and SMYD3 were altered by metabolic cues. They collaboratively supported liver cancer cell proliferation and colony formation. In clinical human liver cancer, both of them were overexpressed. THAP3 positively correlated with OXPHOS gene expression. Together, THAP3 cooperates with SMYD3 to epigenetically upregulate cellular respiration and liver cancer cell proliferation.
Collapse
Affiliation(s)
- Zi-Hao Wang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingyi Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Fuchen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Sijun Sun
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Quan Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, China
| | - Xiaotian Hu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zihan Yin
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Chengmei Xie
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Haiyan Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Tianshi Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, China
| | - Shengjie Zhang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yi-Ping Wang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
15
|
Kopparapu P, Löhr CV, Pearce MC, Tyavanagimatt S, Nakshatri H, Kolluri SK. Small Molecule Functional Converter of B-Cell Lymphoma-2 (Bcl-2) Suppresses Breast Cancer Lung Metastasis. ACS Pharmacol Transl Sci 2024; 7:1302-1309. [PMID: 38751629 PMCID: PMC11091964 DOI: 10.1021/acsptsci.3c00360] [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: 12/13/2023] [Revised: 02/09/2024] [Accepted: 02/28/2024] [Indexed: 05/18/2024]
Abstract
The B-cell lymphoma-2 (Bcl-2) family of proteins plays a vital role in tumorigenesis. Cancer cells utilize the expression of Bcl-2 to evade therapy and develop resistance. Bcl-2 overexpression also causes cancer cells to be more invasive and metastatic. About 80% of cancer deaths are due to metastases, and yet targeted therapies for metastatic cancers are scarce. We discovered a small molecule, BFC1103, which changes the conformation of Bcl-2 to convert the antiapoptotic protein to a proapoptotic protein. BFC1103-induced apoptosis is dependent on the expression levels of Bcl-2, with higher levels causing more apoptosis. BFC1103 suppressed the growth of breast cancer lung metastasis. BFC1103 has the potential for further optimization and development for clinical testing in metastatic cancers that express Bcl-2. This study demonstrates a new approach to target Bcl-2 using a small molecule, BFC1103, to suppress metastatic disease.
Collapse
Affiliation(s)
- Prasad
R. Kopparapu
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331-8580, United States
| | - Christiane V. Löhr
- Department
of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon 97331-4801, United States
| | - Martin C. Pearce
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331-8580, United States
| | - Shanthakumar Tyavanagimatt
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331-8580, United States
| | - Harikrishna Nakshatri
- Department
of Surgery, Indiana University School of
Medicine, Indianapolis, Indiana 46202-3082, United States
| | - Siva K. Kolluri
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331-8580, United States
- Linus
Pauling Institute, Oregon State University, Corvallis, Oregon 97331, United States
| |
Collapse
|
16
|
Çınar İ, Gıdık B, Dirican E. Determination of anti-cancer effects of Nigella sativa seed oil on MCF7 breast and AGS gastric cancer cells. Mol Biol Rep 2024; 51:491. [PMID: 38578469 DOI: 10.1007/s11033-024-09453-1] [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: 01/17/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND This study aimed to investigate the cytotoxic, apoptotic, invasion, metastasis, and heat shock proteins (HSPs) effects of N. sativa oil on breast and gastric cancer cells. METHODS We assessed the cytotoxic and apoptotic effects of various concentrations of N. sativa oil (10-50-100-200 µg/mL) on MCF7 breast cancer and AGS, an adenocarcinoma of the gastric cell line, at 24, 48 and 72 h using the MTT test. Additionally, the expression of the Caspase-3, BCL2/Bax, MMP2-9 and HSP60-70 gene was examined using RT-PCR in cell lines treating with N. sativa. RESULTS The MTT experiments demonstrate that N. sativa has a time and dose-dependent inhibitory effect on the proliferation of MCF7 and AGS cancer cells. The vitality rates of MCF7 and AGS cells treated with N. sativa were 77.04-67.50% at 24 h, 65.28-39.14% at 48 h, and 48.95-32.31% at 72 h. The doses of 100 and 200 µg/mL were shown to be the most effective on both cancer cells. RT-PCR analysis revealed that N. sativa oil extract increased caspase-3 levels in both cell lines at higher concentrations and suppressed BCL2/Bax levels. Exposure of MCF7 and AGS cell lines to N. sativa caused a significant decrease in the expression of MMP2-9 and HSP60-70 genes over time, particularly at a dosage of 200 µg/mL compared to the control group (p < 0.05). CONCLUSIONS Our findings indicate that N. sativa oil has a dose-dependent effect on cytotoxicity and the expression of apoptotic, heat shock proteins, and matrix metalloproteinases genes in breast and gastric cancer.
Collapse
Affiliation(s)
- İrfan Çınar
- Department of Pharmacology, Faculty of Medicine, Kastamonu University, Kastamonu, Turkey
| | - Betül Gıdık
- Department of Organic Farming Management, Bayburt University, Bayburt, 69000, Turkey
| | - Ebubekir Dirican
- Department of Medical Biology, Faculty of Medicine, Bilecik Şeyh Edabali University, Bilecik, Turkey.
| |
Collapse
|
17
|
Wei X, Liu J, Xu Z, Wang D, Zhu Q, Chen Q, Xu W. Research progress on the pharmacological mechanism, in vivo metabolism and structural modification of Erianin. Biomed Pharmacother 2024; 173:116295. [PMID: 38401517 DOI: 10.1016/j.biopha.2024.116295] [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: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
Erianin is an important bibenzyl compound in dendrobium and has a wide spectrum of pharmacological properties. Since Erianin was discovered, abundant results have been achieved in the in vitro synthesis, structural modification, and pharmacological mechanism research. Researchers have developed a series of simple and efficient in vitro synthesis methods to improve the shortcomings of poor water solubility by replacing the chemical structure or coating it in nanomaterials. Erianin has a broad anti-tumor spectrum and significant anti-tumor effects. In addition, Erianin also has pharmacological actions like immune regulation, anti-inflammatory, and anti-angiogenesis. A comprehensive understanding of the synthesis, metabolism, structural modification, and pharmacological action pathways of Erianin is of great value for the utilization of Erianin. Therefore, this review conducts a relatively systematic look back at Erianin from the above four aspects, to give a reference for the evolvement and further appliance of Erianin.
Collapse
Affiliation(s)
- Xin Wei
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Jiajia Liu
- University of Science and Technology of China, Hefei 230026, PR China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, PR China
| | - Ziming Xu
- University of Science and Technology of China, Hefei 230026, PR China; Department of Ophthalmology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, PR China
| | - Dan Wang
- University of Science and Technology of China, Hefei 230026, PR China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, PR China
| | - Qizhi Zhu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Qi Chen
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Weiping Xu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, PR China; Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei 230001, PR China.
| |
Collapse
|
18
|
Zhou X, Qian Y, Ling C, He Z, Shi P, Gao Y, Sui X. An integrated framework for prognosis prediction and drug response modeling in colorectal liver metastasis drug discovery. J Transl Med 2024; 22:321. [PMID: 38555418 PMCID: PMC10981831 DOI: 10.1186/s12967-024-05127-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/23/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most prevalent cancer globally, and liver metastasis (CRLM) is the primary cause of death. Hence, it is essential to discover novel prognostic biomarkers and therapeutic drugs for CRLM. METHODS This study developed two liver metastasis-associated prognostic signatures based on differentially expressed genes (DEGs) in CRLM. Additionally, we employed an interpretable deep learning model utilizing drug sensitivity databases to identify potential therapeutic drugs for high-risk CRLM patients. Subsequently, in vitro and in vivo experiments were performed to verify the efficacy of these compounds. RESULTS These two prognostic models exhibited superior performance compared to previously reported ones. Obatoclax, a BCL-2 inhibitor, showed significant differential responses between high and low risk groups classified by prognostic models, and demonstrated remarkable effectiveness in both Transwell assay and CT26 colorectal liver metastasis mouse model. CONCLUSIONS This study highlights the significance of developing specialized prognostication approaches and investigating effective therapeutic drugs for patients with CRLM. The application of a deep learning drug response model provides a new drug discovery strategy for translational medicine in precision oncology.
Collapse
Affiliation(s)
- Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Yuzhen Qian
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chen Ling
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Zhuoying He
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Peishang Shi
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China.
| | - Xinghua Sui
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China.
| |
Collapse
|
19
|
Chi H, Su L, Yan Y, Gu X, Su K, Li H, Yu L, Liu J, Wang J, Wu Q, Yang G. Illuminating the immunological landscape: mitochondrial gene defects in pancreatic cancer through a multiomics lens. Front Immunol 2024; 15:1375143. [PMID: 38510247 PMCID: PMC10953916 DOI: 10.3389/fimmu.2024.1375143] [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: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
This comprehensive review delves into the complex interplay between mitochondrial gene defects and pancreatic cancer pathogenesis through a multiomics approach. By amalgamating data from genomic, transcriptomic, proteomic, and metabolomic studies, we dissected the mechanisms by which mitochondrial genetic variations dictate cancer progression. Emphasis has been placed on the roles of these genes in altering cellular metabolic processes, signal transduction pathways, and immune system interactions. We further explored how these findings could refine therapeutic interventions, with a particular focus on precision medicine applications. This analysis not only fills pivotal knowledge gaps about mitochondrial anomalies in pancreatic cancer but also paves the way for future investigations into personalized therapy options. This finding underscores the crucial nexus between mitochondrial genetics and oncological immunology, opening new avenues for targeted cancer treatment strategies.
Collapse
Affiliation(s)
- Hao Chi
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yalan Yan
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xiang Gu
- Biology Department, Southern Methodist University, Dallas, TX, United States
| | - Ke Su
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lili Yu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jue Wang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Guanhu Yang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
20
|
Kopparapu PR, Pearce MC, Löhr CV, Duong C, Jang HS, Tyavanagimatt S, O'Donnell EF, Nakshatri H, Kolluri SK. Identification and Characterization of a Small Molecule Bcl-2 Functional Converter. CANCER RESEARCH COMMUNICATIONS 2024; 4:634-644. [PMID: 38329389 PMCID: PMC10911799 DOI: 10.1158/2767-9764.crc-22-0526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/21/2023] [Accepted: 02/06/2024] [Indexed: 02/09/2024]
Abstract
Cancer cells exploit the expression of anti-apoptotic protein Bcl-2 to evade apoptosis and develop resistance to therapeutics. High levels of Bcl-2 leads to sequestration of pro-apoptotic proteins causing the apoptotic machinery to halt. In this study, we report discovery of a small molecule, BFC1108 (5-chloro-N-(2-ethoxyphenyl)-2-[(4-methoxybenzyol)amino]benzamide), which targets Bcl-2 and converts it into a pro-apoptotic protein. The apoptotic effect of BFC1108 is not inhibited, but rather potentiated, by Bcl-2 overexpression. BFC1108 induces a conformational change in Bcl-2, resulting in the exposure of its BH3 domain both in vitro and in vivo. BFC1108 suppresses the growth of triple-negative breast cancer xenografts with high Bcl-2 expression and inhibits breast cancer lung metastasis. This study demonstrates a novel approach to targeting Bcl-2 using BFC1108, a small molecule Bcl-2 functional converter that effectively induces apoptosis in Bcl-2-expressing cancers. SIGNIFICANCE We report the identification of a small molecule that exposes the Bcl-2 killer conformation and induces death in Bcl-2-expressing cancer cells. Selective targeting of Bcl-2 and elimination of cancer cells expressing Bcl-2 opens up new therapeutic avenues.
Collapse
Affiliation(s)
- Prasad R. Kopparapu
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Martin C. Pearce
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Christiane V. Löhr
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon
| | - Cathy Duong
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Hyo Sang Jang
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Shanthakumar Tyavanagimatt
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Edmond F. O'Donnell
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | | | - Siva K. Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
| |
Collapse
|
21
|
Aibinder P, Cohen-Erez I, Rapaport H. Rational Formulation of targeted ABT-737 nanoparticles by self-assembled polypeptides and designed peptides. Heliyon 2024; 10:e26095. [PMID: 38420433 PMCID: PMC10900936 DOI: 10.1016/j.heliyon.2024.e26095] [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: 10/24/2023] [Revised: 12/11/2023] [Accepted: 02/07/2024] [Indexed: 03/02/2024] Open
Abstract
Here we present the development of nanoparticles (NPs) formulations specifically designed for targeting the antiapoptotic Bcl-2 proteins on the outer membrane of mitochondria with the drug agent ABT-737. The NPs which are self-assembled by the natural polypeptide poly gamma glutamic acid (ϒPGA) and a designed cationic and amphiphilic peptide (PFK) have been shown to target drugs toward mitochondria. In this study we systematically developed the formulation of such NPs loaded with the ABT-737 and demonstrated the cytotoxic effect of the best identified formulation on MDA-MB-231 cells. Our findings emphasize the critical role of solutions pH and the charged state of the components throughout the formulation process as well as the concentrations of the co-components and their mixing sequence, in achieving the most stable and effective cytotoxic formulation. Our study highlights the potential versatility of designed peptides in combination with biopolymers for improving drug delivery formulations and enhance their targeting abilities.
Collapse
Affiliation(s)
- Polina Aibinder
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Ifat Cohen-Erez
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Hanna Rapaport
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Ilse Katz Institute for Nanoscale Science and Technology (IKI), Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
22
|
Singh S. Review on Natural Agents as Aromatase Inhibitors: Management of Breast Cancer. Comb Chem High Throughput Screen 2024; 27:2623-2638. [PMID: 37861041 DOI: 10.2174/0113862073269599231009115338] [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: 06/28/2023] [Revised: 08/16/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023]
Abstract
Breast cancer is a prevalent type of cancer that is typically hormone-dependent, caused by estrogen. Aromatase inhibitors are frequently utilised in the treatment of hormonereceptor- positive breast cancer because they prevent the enzyme aromatase from converting androgens to estrogens. Natural medicines with aromatase inhibitory characteristics have attracted interest as potential alternatives or complementary therapy to manufactured medications. This review discusses the function of natural agents as aromatase inhibitors in treating breast cancer. A variety of natural compounds have been investigated for their capacity to inhibit aromatase activity and lower estrogen levels. These agents include resveratrol from red wine and grapes, curcumin from turmeric extract and green teahigh in catechins, and other flavonoids such as genistein, luteolin and quercetin. It has been demonstrated that by decreasing estrogen synthesis, they can slow the growth of breast cancer cells that are dependent on estrogen. However, the clinical evidence supporting their efficacy and safety in breast cancer treatment is inadequate. More research is required to investigate the therapeutic potential of natural medicines, such as aromatase inhibitors, in treating breast cancer. The clinical trials are required to assess their efficacy, appropriate doses, and potential interactions with other therapies. In conclusion, natural aromatase inhibitory drugs are promising adjuncts in the treatment of hormone receptor-positive breast cancer. Their clinical value and safety profile, however, require additional investigation.
Collapse
Affiliation(s)
- Sonia Singh
- Institute of Pharmaceutical Research, GLA University Mathura, U.P: 281406, India
| |
Collapse
|
23
|
Amiri S, Pashizeh F, Moeinabadi-Bidgoli K, Eyvazi Y, Akbari T, Salehi Moghaddam Z, Eskandarisani M, Farahmand F, Hafezi Y, Nouri Jevinani H, Seif M, Mousavi-Niri N, Chiani M, Tavakkoli Yaraki M. Co-encapsulation of hydrophilic and hydrophobic drugs into niosomal nanocarrier for enhanced breast cancer therapy: In silico and in vitro studies. ENVIRONMENTAL RESEARCH 2023; 239:117292. [PMID: 37806480 DOI: 10.1016/j.envres.2023.117292] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
Combination therapy has been considered one of the most promising approaches for improving the therapeutic effects of anticancer drugs. This is the first study that uses two different antioxidants in full-characterized niosomal formulation and thoroughly evaluates their synergistic effects on breast cancer cells. In this study, in-silico studies of hydrophilic and hydrophobic drugs (ascorbic acid: Asc and curcumin: Cur) interactions and release were investigated and validated by a set of in vitro experiments to reveal the significant improvement in breast cancer therapy using a co-delivery approach by niosomal nanocarrier. The niosomal nanoparticles containing surfactants (Span 60 and Tween 60) and cholesterol at 2:1 M ratio were prepared through the film hydration method. A systematic evaluation of nanoniosomes was carried out. The release profile demonstrated two phases (initial burst followed by sustained release) and a pH-dependent release schedule over 72 h. The optimized niosomal preparation displayed superior storage stability for up to 2 months at 4 °C, exhibiting extremely minor changes in pharmaceutical encapsulation efficiency and size. Free dual drugs (Asc + Cur) and dual-drug loaded niosomes (Niosomal (Asc + Cur)) enhanced the apoptotic activity and cytotoxicity and inhibited cell migration which confirmed the synergistic effect of co-encapsulated drugs. Also, significant up-regulation of p53 and Bax genes was observed in cells treated with Asc + Cur and Niosomal (Asc + Cur), while the anti-apoptotic Bcl-2 gene was down-regulated. These results were in correlation with the increase in the enzyme activity of SOD, CAT, and caspase, and the levels of malondialdehyde (MDA) and reactive oxygen species (ROS) upon treatment with the mentioned drugs. Furthermore, these anti-cancer effects were higher when using Niosomal (Asc + Cur) than Asc + Cur. Histopathological examination also revealed that Niosomal (Asc + Cur) had a lower mitosis index, invasion, and pleomorphism than Asc + Cur. These findings indicated that niosomal formulation for co-delivery of Asc and Cur would offer a promising delivery system for an effective breast cancer treatment.
Collapse
Affiliation(s)
- Sahar Amiri
- Department of Genetic, Islamic Azad University, Tehran North Branch, Iran
| | - Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science Yazd, Iran
| | - Kasra Moeinabadi-Bidgoli
- Departments of Medicine and Endocrinology, University of California San Francisco and San Francisco Veterans Affairs Health Center, San Francisco, CA, USA
| | - Yalda Eyvazi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Tanin Akbari
- Department of Medical Science, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi Moghaddam
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, Iran
| | | | - Faranak Farahmand
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yousef Hafezi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hoda Nouri Jevinani
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdi Seif
- Faculty of Materials Science and Engineering, K.N. Toosi University of Technology, Tehran, Iran
| | - Neda Mousavi-Niri
- Department of Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohsen Chiani
- Department of Nano Biotechnology, New Technology Research Group, Pasteur Institute of Iran, Tehran, Iran.
| | - Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, NSW 2109, Australia.
| |
Collapse
|
24
|
Qin N, Paisana E, Picard D, Leprivier G, Langini M, Custódia C, Cascão R, Conrad C, Peitzsch M, Stefanski A, Stühler K, Fischer U, Faria CC, Dietrich S, Reifenberger G, Remke M. The long non-coding RNA OTX2-AS1 promotes tumor growth and predicts response to BCL-2 inhibition in medulloblastoma. J Neurooncol 2023; 165:329-342. [PMID: 37976029 PMCID: PMC10689561 DOI: 10.1007/s11060-023-04508-y] [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: 10/06/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE Primary brain tumors are a leading cause of cancer-related death in children, and medulloblastoma is the most common malignant pediatric brain tumor. The current molecular characterization of medulloblastoma is mainly based on protein-coding genes, while little is known about the involvement of long non-coding RNAs (lncRNAs). This study aimed to elucidate the role of the lncRNA OTX2-AS1 in medulloblastoma. METHODS Analyses of DNA copy number alterations, methylation profiles, and gene expression data were used to characterize molecular alterations of OTX2-AS1 in medulloblastoma tissue samples. In vitro analyses of medulloblastoma cell models and orthotopic in vivo experiments were carried out for functional characterization of OTX2-AS1. High-throughput drug screening was employed to identify pharmacological inhibitors, while proteomics and metabolomics analyses were performed to address potential mechanisms of drug action. RESULTS We detected amplification and consecutive overexpression of OTX2 and OTX2-AS1 in a subset of medulloblastomas. In addition, OTX2-AS1 promoter methylation was linked to OTX2-AS1 expression. OTX2-AS1 knockout reduced medulloblastoma cell viability and cell migration in vitro and prolonged survival in the D283 orthotopic medulloblastoma mouse xenograft model. Pharmacological inhibition of BCL-2 suppressed the growth of OTX2-AS1 overexpressing medulloblastoma cells in vitro. CONCLUSIONS Our study revealed a pro-tumorigenic role of OTX2-AS1 in medulloblastoma and identified BCL-2 inhibition as a potential therapeutic approach to target OTX2-AS1 overexpressing medulloblastoma cells.
Collapse
Affiliation(s)
- Nan Qin
- Department of Hematology, Oncology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany.
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany.
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University, and University Hospital Düsseldorf, Düsseldorf, Germany.
- High-Throughput Drug Screening Core Facility, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Düsseldorf, Germany.
| | - Eunice Paisana
- Instituto de Medicina Molecular João Lobo Antunes (iMM), Faculdade de Medicina da Universidade de Lisboa, Lisbon, 1649-028, Portugal
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University, and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Gabriel Leprivier
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University, and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Maike Langini
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty Carl Gustav Carus, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Molecular Proteomics Laboratory, Biological and Medical Research Center (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Carlos Custódia
- Instituto de Medicina Molecular João Lobo Antunes (iMM), Faculdade de Medicina da Universidade de Lisboa, Lisbon, 1649-028, Portugal
| | - Rita Cascão
- Instituto de Medicina Molecular João Lobo Antunes (iMM), Faculdade de Medicina da Universidade de Lisboa, Lisbon, 1649-028, Portugal
| | - Catleen Conrad
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty Carl Gustav Carus, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty Carl Gustav Carus, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory, Biological and Medical Research Center (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Molecular Medicine 1, Heinrich Heine University Medical Faculty, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biological and Medical Research Center (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Molecular Medicine 1, Heinrich Heine University Medical Faculty, Düsseldorf, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Claudia C Faria
- Instituto de Medicina Molecular João Lobo Antunes (iMM), Faculdade de Medicina da Universidade de Lisboa, Lisbon, 1649-028, Portugal
- Department of Neurosurgery, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, EPE, Lisbon, 1649-028, Portugal
| | - Sascha Dietrich
- Department of Hematology, Oncology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Guido Reifenberger
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University, and University Hospital Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Marc Remke
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University, and University Hospital Düsseldorf, Düsseldorf, Germany
- High-Throughput Drug Screening Core Facility, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center of Saarland, Homburg/Saar, Germany
| |
Collapse
|
25
|
He X, Lin F, Jia R, Xia Y, Liang Z, Xiao X, Hu Q, Deng X, Li Q, Sheng W. Coordinated modulation of long non-coding RNA ASBEL and curcumin co-delivery through multicomponent nanocomplexes for synchronous triple-negative breast cancer theranostics. J Nanobiotechnology 2023; 21:397. [PMID: 37904215 PMCID: PMC10617238 DOI: 10.1186/s12951-023-02168-8] [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: 08/02/2023] [Accepted: 10/18/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Abnormally regulated long non-coding RNAs (lncRNAs) functions in cancer emphasize their potential to serve as potential targets for cancer therapeutic intervention. LncRNA ASBEL has been identified as oncogene and an anti-sense transcript of tumor-suppressor gene of BTG3 in triple-negative breast cancer (TNBC). RESULTS Herein, multicomponent self-assembled polyelectrolyte nanocomplexes (CANPs) based on the polyelectrolytes of bioactive hyaluronic acid (HA) and chitosan hydrochloride (CS) were designed and prepared for the collaborative modulation of oncogenic lncRNA ASBEL with antago3, an oligonucleotide antagonist targeting lncRNA ASBEL and hydrophobic curcumin (Cur) co-delivery for synergetic TNBC therapy. Antago3 and Cur co-incorporated CANPs were achieved via a one-step assembling strategy with the cooperation of noncovalent electrostatic interactions, hydrogen-bonding, and hydrophobic interactions. Moreover, the multicomponent assembled CANPs were ulteriorly decorated with a near-infrared fluorescence (NIRF) Cy-5.5 dye (FCANPs) for synchronous NIRF imaging and therapy monitoring performance. Resultantly, MDA-MB-231 cells proliferation, migration, and invasion were efficiently inhibited, and the highest apoptosis ratio was induced by FCANPs with coordination patterns. At the molecular level, effective regulation of lncRNA ASBEL/BTG3 and synchronous regulation of Bcl-2 and c-Met pathways could be observed. CONCLUSION As expected, systemic administration of FCANPs resulted in targeted and preferential accumulation of near-infrared fluorescence signal and Cur in the tumor tissue. More attractively, systemic FCANPs-mediated collaborative modulating lncRNA ASBEL/BTG3 and Cur co-delivery significantly suppressed the MDA-MB-231 xenograft tumor growth, inhibited metastasis and extended survival rate with negligible systemic toxicity. Our present study represented an effective approach to developing a promising theranostic platform for combating TNBC in a combined therapy pattern.
Collapse
Affiliation(s)
- Xuesong He
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Fengjuan Lin
- Department of Oncology, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Runqing Jia
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Yang Xia
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Zhaoyuan Liang
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Xiangqian Xiao
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Qin Hu
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Xiongwei Deng
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China.
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, 100049, China.
| | - Qun Li
- Department of Oncology, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200123, China.
| | - Wang Sheng
- Department of Environment and Life Science, Beijing International Science and Technology, Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China.
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, 100049, China.
| |
Collapse
|
26
|
Mohamed EE, Ahmed OM, Zoheir KMA, El-Shahawy AAG, Tamur S, Shams A, Burcher JT, Bishayee A, Abdel-Moneim A. Naringin-Dextrin Nanocomposite Abates Diethylnitrosamine/Acetylaminofluorene-Induced Lung Carcinogenesis by Modulating Oxidative Stress, Inflammation, Apoptosis, and Cell Proliferation. Cancers (Basel) 2023; 15:5102. [PMID: 37894468 PMCID: PMC10605195 DOI: 10.3390/cancers15205102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Nanotechnology has proven advantageous in numerous scientific applications, one being to enhance the delivery of chemotherapeutic agents. This present study aims to evaluate the mechanisms underlying the chemopreventive action of naringin-dextrin nanocomposites (Nar-Dx-NCs) against diethylnitrosamine (DEN)/2-acetylaminofluorene (2AAF)-induced lung carcinogenesis in male Wistar rats. DEN was administered intraperitoneally (i.p.) (150 mg/kg/week) for two weeks, followed by the oral administration of 2AAF (20 mg/kg) four times a week for three weeks. Rats receiving DEN/2AAF were concurrently treated with naringin or Nar-Dx-NCs orally at a dose of 10 mg/kg every other day for 24 weeks. Naringin and Nar-Dx-NCs treatments prevented the formation of tumorigenic cells within the alveoli of rats exposed to DEN/2AAF. These findings were associated with a significant decrease in lipid peroxidation, upregulation of antioxidant enzyme (glutathione peroxidase and superoxide dismutase) activity, and enhanced glutathione and nuclear factor erythroid 2-related factor 2 expression in the lungs. Naringin and Nar-Dx-NCs exerted anti-inflammatory actions manifested by a decrease in lung protein expression of tumor necrosis factor-α and interleukin-1β and mRNA expression of interleukin-6, interferon-γ, nuclear factor-κB, and inducible nitric oxide synthase, with a concurrent increase in interleukin-10 expression. The anti-inflammatory effect of Nar-Dx-NCs was more potent than naringin. Regarding the effect on apoptosis, both naringin and Nar-Dx-NCs significantly reduced Bcl-2 and increased Bax and P53 expressions. Moreover, naringin or Nar-Dx-NCs induced a significant decrease in the expression of the proliferator marker, Ki-67, and the effect of Nar-Dx-NCs was more marked. In conclusion, Nar-Dx-NCs improved naringin's preventive action against DEN/2AAF-induced lung cancer and exerted anticarcinogenic effects by suppressing oxidative stress and inflammation and improving apoptotic signal induction and propagation.
Collapse
Affiliation(s)
- Eman E. Mohamed
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| | - Osama M. Ahmed
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| | - Khairy M. A. Zoheir
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Cairo 12622, Egypt;
| | - Ahmed A. G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Shadi Tamur
- Departement of Pediatrics, College of Medicine, Taif University, Taif 21944, Saudi Arabia;
| | - Anwar Shams
- Departement of Pharmacology, College of Medicine, Taif University, Taif 21944, Saudi Arabia
- Centre of Biomedical Sciences Research, Deanship of Scientific Research, Taif University, Taif 21974, Saudi Arabia
- High Altitude Research Center, Taif University, Taif 21944, Saudi Arabia
| | - Jack T. Burcher
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.T.B.); or (A.B.)
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (J.T.B.); or (A.B.)
| | - Adel Abdel-Moneim
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (E.E.M.)
| |
Collapse
|
27
|
Gupta SRR, Mittal P, Kundu B, Singh A, Singh IK. Silibinin: an inhibitor for a high-expressed BCL-2A1/BFL1 protein, linked with poor prognosis in breast cancer. J Biomol Struct Dyn 2023:1-11. [PMID: 37837418 DOI: 10.1080/07391102.2023.2268176] [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: 03/14/2023] [Accepted: 09/29/2023] [Indexed: 10/16/2023]
Abstract
Breast cancer (BC) accounts for 30% of all diagnosed cases of cancer in women and remains a leading cause of cancer-related deaths among women worldwide. The current study looks for a protein from the anti-apoptotic/pro-survival BCL-2 family whose overexpression reduces survivability in BC patients and a potential inhibitor for the protein. We found BCL-2A1/BFL1 protein with high expression linked to low survivability in BC. The protein shows prognosis in 8 out of 29 categories, whereas no other family member manifests this property. Out of 7379 compounds, three small molecules (CHEMBL9509, CHEMBL2104550 and CHEMBL3545011) form an H-bond with BCL-2A1/BFL1 protein's unique residue Cys55. Of the three small molecules, we found CHEMBL9509 (Silibinin) to be a potent inhibitor. The compound forms a stable H-bond with the residue Cys55 with the lowest binding energy compared to the other two compounds. It remains stable in the BH3 binding region for more than 100 ns, whereas the other two detach from the region. Additionally, the compound is found to be better than Venetoclax and Nematoclax. We firmly believe in the compound CHEMBL9509 potency to halt BC's progression by inhibiting the BCL-2A1/BFL1 protein, increasing patients' survivability.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shradheya R R Gupta
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
| | - Pooja Mittal
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
- Norris Comprehensive Cancer Center, Division of Medical Oncology, University of Southern California, Los Angeles, USA
| | - Bishwajit Kundu
- Kusuma School of Biological Science, Indian Institute of Technology Delhi, New Delhi, India
| | - Archana Singh
- Department of Plant Molecular Biology, University of Delhi (South Campus), New Delhi, India
| | - Indrakant K Singh
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
- Norris Comprehensive Cancer Center, Division of Medical Oncology, University of Southern California, Los Angeles, USA
- Institute of Eminence, Delhi School of Public Health, University of Delhi, Delhi, India
| |
Collapse
|
28
|
Almehdi AM, Soliman SSM, El-Shorbagi ANA, Westwell AD, Hamdy R. Design, Synthesis, and Potent Anticancer Activity of Novel Indole-Based Bcl-2 Inhibitors. Int J Mol Sci 2023; 24:14656. [PMID: 37834104 PMCID: PMC10572575 DOI: 10.3390/ijms241914656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 10/15/2023] Open
Abstract
The Bcl-2 family plays a crucial role in regulating cell apoptosis, making it an attractive target for cancer therapy. In this study, a series of indole-based compounds, U1-6, were designed, synthesized, and evaluated for their anticancer activity against Bcl-2-expressing cancer cell lines. The binding affinity, safety profile, cell cycle arrest, and apoptosis effects of the compounds were tested. The designed compounds exhibited potent inhibitory activity at sub-micromolar IC50 concentrations against MCF-7, MDA-MB-231, and A549 cell lines. Notably, U2 and U3 demonstrated the highest activity, particularly against MCF-7 cells. Respectively, both U2 and U3 showed potential BCL-2 inhibition activity with IC50 values of 1.2 ± 0.02 and 11.10 ± 0.07 µM using an ELISA binding assay compared with 0.62 ± 0.01 µM for gossypol, employed as a positive control. Molecular docking analysis suggested stable interactions of compound U2 at the Bcl-2 binding site through hydrogen bonding, pi-pi stacking, and hydrophobic interactions. Furthermore, U2 demonstrated significant induction of apoptosis and cell cycle arrest at the G1/S phase. Importantly, U2 displayed a favourable safety profile on HDF human dermal normal fibroblast cells at 10-fold greater IC50 values compared with MDA-MB-231 cells. These findings underscore the therapeutic potential of compound U2 as a Bcl-2 inhibitor and provide insights into its molecular mechanisms of action.
Collapse
Affiliation(s)
- Ahmed M. Almehdi
- College of Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
- Research Institute for Science and Engineering (RISE), University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Sameh S. M. Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
| | | | - Andrew D. Westwell
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff CF10 3NB, UK
| | - Rania Hamdy
- Research Institute for Science and Engineering (RISE), University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
- Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
29
|
MALIK MUNEEBA, MAQBOOL MAMOONA, NISAR TOOBA, AKHTER TAZEEM, UJAN JAVEDAHMED, ALGARNI ALANOODS, JOUFI FAKHRIAAAL, ALANAZI SULTANSHAFIK, ALMOTARED MOHAMMADHADI, BEKHIT MOUNIRMSALEM, JAMIL MUHAMMAD. Deciphering key genes involved in cisplatin resistance in kidney renal clear cell carcinoma through a combined in silico and in vitro approach. Oncol Res 2023; 31:899-916. [PMID: 37744271 PMCID: PMC10513959 DOI: 10.32604/or.2023.030760] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/29/2023] [Indexed: 09/26/2023] Open
Abstract
The low survival rate of Kidney renal clear cell carcinoma (KIRC) patients is largely attributed to cisplatin resistance. Rather than focusing solely on individual proteins, exploring protein-protein interactions could offer greater insight into drug resistance. To this end, a series of in silico and in vitro experiments were conducted to identify hub genes in the intricate network of cisplatin resistance-related genes in KIRC chemotherapy. The genes involved in cisplatin resistance across KIRC were retrieved from the National Center for Biotechnology Information (NCBI) database using search terms as "Kidney renal clear cell carcinoma" and "Cisplatin resistance". The genes retrieved were analyzed for hub gene identification using the STRING database and Cytoscape tool. Expression and promoter methylation profiling of the hub genes was done using UALCAN, GEPIA, OncoDB, and HPA databases. Mutational, survival, functional enrichment, immune cell infiltration, and drug prediction analyses of the hub genes were performed using the cBioPortal, GEPIA, GSEA, TIMER, and DrugBank databases. Lastly, expression and methylation levels of the hub genes were validated on two cisplatin-resistant RCC cell lines (786-O and A-498) and a normal renal tubular epithelial cell line (HK-2) using two high throughput techniques, including targeted bisulfite sequencing (bisulfite-seq) and RT-qPCR. A total of 124 genes were identified as being associated with cisplatin resistance in KIRC. Out of these genes, MCL1, IGF1R, CCND1, and PTEN were identified as hub genes and were found to have significant (p < 0.05) variations in their mRNA and protein expressions and effects on the overall survival (OS) of the KIRC patients. Moreover, an aberrant promoter methylation pattern was found to be associated with the dysregulation of the hub genes. In addition to this, hub genes were also linked with different cisplatin resistance-causing pathways. Thus, hub genes can be targeted with Alvocidib, Estradiol, Tretinoin, Capsaicin, Dronabinol, Metribolone, Calcitriol, Acetaminophen, Acitretin, Cyclosporine, Azacitidine, Genistein, and Resveratrol drugs. As the pathogenesis of KIRC is complex, targeting hub genes and associated pathways involved in cisplatin resistance could bring a milestone change in the drug discovery and management of drug resistance, which might uplift overall survival among KIRC patients.
Collapse
Affiliation(s)
| | | | | | - TAZEEM AKHTER
- Public Health Department, University of Health Sciences, Lahore, Pakistan
| | - JAVED AHMED UJAN
- Department of Zoology, Shah Abdul Latif University, Khairpur, Pakistan
- Department of Animal Sciences, University of Florida, Gainesville, USA
| | - ALANOOD S. ALGARNI
- Pharmacology and Toxicology Department College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - FAKHRIA A. AL JOUFI
- Department of Pharmacology, College of Pharmacy, Jouf University, Aljouf, Saudi Arabia
| | | | | | - MOUNIR M. SALEM BEKHIT
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - MUHAMMAD JAMIL
- PARC Arid Zone Research Center, Dera Ismail Khan, Pakistan
| |
Collapse
|
30
|
Melotti L, Venerando A, Zivelonghi G, Carolo A, Marzorati S, Martinelli G, Sugni M, Maccatrozzo L, Patruno M. A Second Life for Seafood Waste: Therapeutical Promises of Polyhydroxynapthoquinones Extracted from Sea Urchin by-Products. Antioxidants (Basel) 2023; 12:1730. [PMID: 37760033 PMCID: PMC10526080 DOI: 10.3390/antiox12091730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Coping with a zero-waste, more sustainable economy represents the biggest challenge for food market nowadays. We have previously demonstrated that by applying smart multidisciplinary waste management strategies to purple sea urchin (Paracentrotus lividus) food waste, it is possible to obtain both a high biocompatible collagen to produce novel skin substitutes and potent antioxidant pigments, namely polyhydroxynapthoquinones (PHNQs). Herein, we have analyzed the biological activities of the PHNQs extract, composed of Spinochrome A and B, on human skin fibroblast cells to explore their future applicability in the treatment of non-healing skin wounds with the objective of overcoming the excessive oxidative stress that hinders wound tissue regeneration. Our results clearly demonstrate that the antioxidant activity of PHNQs is not restricted to their ability to scavenge reactive oxygen species; rather, it can be traced back to an upregulating effect on the expression of superoxide dismutase 1, one of the major components of the endogenous antioxidant enzymes defense system. In addition, the PHNQs extract, in combination with Antimycin A, displayed a synergistic pro-apoptotic effect, envisaging its possible employment against chemoresistance in cancer treatments. Overall, this study highlights the validity of a zero-waste approach in the seafood chain to obtain high-value products, which, in turn, may be exploited for different biomedical applications.
Collapse
Affiliation(s)
- Luca Melotti
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, 35020 Padova, Italy; (L.M.); (G.Z.); (A.C.); (L.M.); (M.P.)
| | - Andrea Venerando
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Via delle Scienze 206, 33100 Udine, Italy
| | - Giulia Zivelonghi
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, 35020 Padova, Italy; (L.M.); (G.Z.); (A.C.); (L.M.); (M.P.)
| | - Anna Carolo
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, 35020 Padova, Italy; (L.M.); (G.Z.); (A.C.); (L.M.); (M.P.)
| | - Stefania Marzorati
- Department of Environmental Science and Policy, University of Milan, Via Celoria 2, 20133 Milan, Italy; (S.M.); (G.M.); (M.S.)
| | - Giordana Martinelli
- Department of Environmental Science and Policy, University of Milan, Via Celoria 2, 20133 Milan, Italy; (S.M.); (G.M.); (M.S.)
| | - Michela Sugni
- Department of Environmental Science and Policy, University of Milan, Via Celoria 2, 20133 Milan, Italy; (S.M.); (G.M.); (M.S.)
| | - Lisa Maccatrozzo
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, 35020 Padova, Italy; (L.M.); (G.Z.); (A.C.); (L.M.); (M.P.)
| | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, 35020 Padova, Italy; (L.M.); (G.Z.); (A.C.); (L.M.); (M.P.)
| |
Collapse
|
31
|
González-Arzola K, Díaz-Quintana A. Mitochondrial Factors in the Cell Nucleus. Int J Mol Sci 2023; 24:13656. [PMID: 37686461 PMCID: PMC10563088 DOI: 10.3390/ijms241713656] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
The origin of eukaryotic organisms involved the integration of mitochondria into the ancestor cell, with a massive gene transfer from the original proteobacterium to the host nucleus. Thus, mitochondrial performance relies on a mosaic of nuclear gene products from a variety of genomes. The concerted regulation of their synthesis is necessary for metabolic housekeeping and stress response. This governance involves crosstalk between mitochondrial, cytoplasmic, and nuclear factors. While anterograde and retrograde regulation preserve mitochondrial homeostasis, the mitochondria can modulate a wide set of nuclear genes in response to an extensive variety of conditions, whose response mechanisms often merge. In this review, we summarise how mitochondrial metabolites and proteins-encoded either in the nucleus or in the organelle-target the cell nucleus and exert different actions modulating gene expression and the chromatin state, or even causing DNA fragmentation in response to common stress conditions, such as hypoxia, oxidative stress, unfolded protein stress, and DNA damage.
Collapse
Affiliation(s)
- Katiuska González-Arzola
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Consejo Superior de Investigaciones Científicas—Universidad de Sevilla—Universidad Pablo de Olavide, 41092 Seville, Spain
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, 41012 Seville, Spain
| | - Antonio Díaz-Quintana
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, 41012 Seville, Spain
- Instituto de Investigaciones Químicas—cicCartuja, Universidad de Sevilla—C.S.I.C, 41092 Seville, Spain
| |
Collapse
|
32
|
Thiruvengadam R, Kim JH. Therapeutic strategy for oncovirus-mediated oral cancer: A comprehensive review. Biomed Pharmacother 2023; 165:115035. [PMID: 37364477 DOI: 10.1016/j.biopha.2023.115035] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/02/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Oral cancer is a neoplastic disorder of the oral cavities, including the lips, tongue, buccal mucosa, and lower and upper gums. Oral cancer assessment entails a multistep process that requires deep knowledge of the molecular networks involved in its progression and development. Preventive measures including public awareness of risk factors and improving public behaviors are necessary, and screening techniques should be encouraged to enable early detection of malignant lesions. Herpes simplex virus (HSV), human papillomavirus (HPV), Epstein-Barr virus (EBV), and Kaposi sarcoma-associated herpesvirus (KSHV) are associated with other premalignant and carcinogenic conditions leading to oral cancer. Oncogenic viruses induce chromosomal rearrangements; activate signal transduction pathways via growth factor receptors, cytoplasmic protein kinases, and DNA binding transcription factors; modulate cell cycle proteins, and inhibit apoptotic pathways. In this review, we present an up-to-date overview on the use of nanomaterials for regulating viral proteins and oral cancer as well as the role of phytocompounds on oral cancer. The targets linking oncoviral proteins and oral carcinogenesis were also discussed.
Collapse
Affiliation(s)
- Rekha Thiruvengadam
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea.
| |
Collapse
|
33
|
Saygin C, Giordano G, Shimamoto K, Eisfelder B, Thomas-Toth A, Venkataraman G, Ananthanarayanan V, Vincent TL, DuVall A, Patel AA, Chen Y, Tan F, Anthony SP, Chen Y, Shen Y, Odenike O, Teachey DT, Kee BL, LaBelle J, Stock W. Dual Targeting of Apoptotic and Signaling Pathways in T-Lineage Acute Lymphoblastic Leukemia. Clin Cancer Res 2023; 29:3151-3161. [PMID: 37363966 PMCID: PMC10425730 DOI: 10.1158/1078-0432.ccr-23-0415] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/08/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
PURPOSE Relapsed T-acute lymphoblastic leukemia (T-ALL) has limited treatment options. We investigated mechanisms of resistance to BH3 mimetics in T-ALL to develop rational combination strategies. We also looked at the preclinical efficacy of NWP-0476, a novel BCL-2/BCL-xL inhibitor, as single agent and combination therapy in T-ALL. EXPERIMENTAL DESIGN We used BH3 profiling as a predictive tool for BH3 mimetic response in T-ALL. Using isogenic control, venetoclax-resistant (ven-R) and NWP-0476-resistant (NWP-R) cells, phosphokinase array was performed to identify differentially regulated signaling pathways. RESULTS Typical T-ALL cells had increased dependence on BCL-xL, whereas early T-precursor (ETP)-ALL cells had higher BCL-2 dependence for survival. BCL-2/BCL-xL dual inhibitors were effective against both subtypes of T-lineage ALL. A 71-protein human phosphokinase array showed increased LCK activity in ven-R cells, and increased ACK1 activity in ven-R and NWP-R cells. We hypothesized that pre-TCR and ACK1 signaling pathways are drivers of resistance to BCL-2 and BCL-xL inhibition, respectively. First, we silenced LCK gene in T-ALL cell lines, which resulted in increased sensitivity to BCL-2 inhibition. Mechanistically, LCK activated NF-κB pathway and the expression of BCL-xL. Silencing ACK1 gene resulted in increased sensitivity to both BCL-2 and BCL-xL inhibitors. ACK1 signaling upregulated AKT pathway, which inhibited the pro-apoptotic function of BAD. In a T-ALL patient-derived xenograft model, combination of NWP-0476 and dasatinib demonstrated synergy without major organ toxicity. CONCLUSIONS LCK and ACK1 signaling pathways are critical regulators of BH3 mimetic resistance in T-ALL. Combination of BH3 mimetics with tyrosine kinase inhibitors might be effective against relapsed T-ALL.
Collapse
Affiliation(s)
- Caner Saygin
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Giorgia Giordano
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Kathryn Shimamoto
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Bart Eisfelder
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | | | | | | | - Tiffaney L. Vincent
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Adam DuVall
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Anand A. Patel
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Yi Chen
- Newave Pharmaceutical Inc., Pleasanton, California
| | - Fenlai Tan
- Newave Pharmaceutical Inc., Pleasanton, California
| | | | - Yu Chen
- Newave Pharmaceutical Inc., Pleasanton, California
| | - Yue Shen
- Newave Pharmaceutical Inc., Pleasanton, California
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - David T. Teachey
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Barbara L. Kee
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - James LaBelle
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Wendy Stock
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
34
|
Ilhan S, Çamli Pulat Ç, Oguz F, Bektaş H, Menteşe E, Atmaca H. Design and synthesis of benzimidazole derivatives as apoptosis-inducing agents by targeting Bcl-2 protein. Mol Divers 2023; 27:1703-1712. [PMID: 36065037 DOI: 10.1007/s11030-022-10524-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/26/2022] [Indexed: 11/24/2022]
Abstract
Bcl-2, an anti-apoptotic protein, is a well-known and appealing cancer therapy target. Novel series of benzimidazole derivatives were synthesized and tested for their activity as Bcl-2 inhibitors on T98G glioblastoma, PC3 prostate, MCF-7 breast, and H69AR lung cancer cells. MTT assay was used to evaluate the cytotoxic effect. PI Annexin V Apoptosis Detection Kit was used to detect apoptosis. Expression levels of the Bcl-2 protein were examined by the Western blot analysis and qRT-PCR. All synthesized benzimidazole derivatives exhibited a cytotoxic effect on cancer cells with IC50 values in the range of 25.2-88.2 µg/mL. Among all derivatives, compounds C1 and D1 demonstrated a higher cytotoxic effect on cancer cells with IC50 values < 50 µg/mL, while a lower cytotoxic effect against human embryonic kidney cells with IC50 values of > 100 µg/mL. C1 and D1 caused a significant increase in the percentage of apoptotic cells in all types of cancer cell cells and both Bcl-2 mRNA and protein levels were significantly reduced. These results suggest that the novel benzimidazole derivatives may be candidates for apoptosis-inducing agents in cancer treatment by targeting anti-Bcl-2 proteins in cancer cells.
Collapse
Affiliation(s)
- Suleyman Ilhan
- Department of Biology, Faculty of Science and Letters, Celal Bayar University, 45140, Manisa, Turkey
| | - Çisil Çamli Pulat
- Applied Science Research Center, Manisa Celal Bayar University, Manisa, Turkey
| | - Ferdi Oguz
- Department of Biology, Faculty of Science and Letters, Celal Bayar University, 45140, Manisa, Turkey
| | - Hakan Bektaş
- Department of Chemistry, Faculty of Science and Art, Giresun University, Giresun, Turkey
| | - Emre Menteşe
- Department of Chemistry, Faculty of Science and Art, Recep Tayyip Erdogan University, Rize, Turkey
| | - Harika Atmaca
- Department of Biology, Faculty of Science and Letters, Celal Bayar University, 45140, Manisa, Turkey.
| |
Collapse
|
35
|
Dong S, Matossian MD, Yousefi H, Khosla M, Collins-Burow BM, Burow ME, Alahari SK. Targeting Mcl-1 by a small molecule NSC260594 for triple-negative breast cancer therapy. Sci Rep 2023; 13:11843. [PMID: 37481672 PMCID: PMC10363135 DOI: 10.1038/s41598-023-37058-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/15/2023] [Indexed: 07/24/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) are aggressive forms of breast cancer and tend to grow and spread more quickly than most other types of breast cancer. TNBCs can neither be targeted by hormonal therapies nor the antibody trastuzumab that targets the HER2 protein. There are urgent unmet medical needs to develop targeted drugs for TNBCs. We identified a small molecule NSC260594 from the NCI diversity set IV compound library. NSC260594 exhibited dramatic cytotoxicity in multiple TNBCs in a dose-and time-dependent manner. NSC260594 inhibited the Myeloid cell leukemia-1 (Mcl-1) expression through downregulation of Wnt signaling proteins. Consistent with this, NSC260594 treatment increased apoptosis, which was confirmed by using an Annexin-V/PI assay. Interestingly, NSC260594 treatment reduced the cancer stem cell (CSC) population in TNBCs. To make NSC260594 more clinically relevant, we treated NSC260594 with TNBC cell derived xenograft (CDX) mouse model, and with patient-derived xenograft (PDX) organoids. NSC260594 significantly suppressed MDA-MB-231 tumor growth in vivo, and furthermore, the combination treatment of NSC260594 and everolimus acted synergistically to decrease growth of TNBC PDX organoids. Together, we found that NSC260594 might serve as a lead compound for triple-negative breast cancer therapy through targeting Mcl-1.
Collapse
Affiliation(s)
- Shengli Dong
- TYK Medicines Inc., Block D, No. 778 Huaxi Avenue, Changxing, 313100, Zhejiang, People's Republic of China.
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA.
| | | | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA
| | - Maninder Khosla
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA
| | | | - Matthew E Burow
- Tulane University School of Medicine, New Orleans, LA, 70118, USA
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC, CSRB 406, New Orleans, LA, 70112, USA.
- Stanley S. Scott Cancer Center, LSUHSC School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
36
|
Lee D, Lee S, Jang YS, Ryoo R, Kim JK, Kang KS, Kim KH. N, N-Dimethyl-anthranilic Acid from Calvatia nipponica Mushroom Fruiting Bodies Induces Apoptotic Effects on MDA-MB-231 Human Breast Cancer Cells. Nutrients 2023; 15:3091. [PMID: 37513511 PMCID: PMC10386113 DOI: 10.3390/nu15143091] [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: 06/19/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer ranks among the most prevalent malignancies affecting women worldwide, and apoptosis-targeting drugs are attractive candidates for the treatment of cancer. In the current study, we investigated the in vitro cytotoxicity of the mushroom Calvatia nipponica in human breast cancer cells (MDA-MB-231), identified potential antitumor compounds through bioactivity-guided isolation, and elucidated the antitumor, pro-apoptotic molecular mechanisms of the identified bioactive compounds. C. nipponica is edible when young, and it has been used as a food source as well as a traditional medicine in wound dressings. However, only a limited number of studies have reported its chemical composition and biological activities. In the screening test, the methanol extract of C. nipponica fruiting bodies exhibited cytotoxicity against MDA-MB-231 cells. Bioactivity-guided fractionation of the methanol (MeOH) extract and chemical investigation of the active fractions resulted in the isolation of fourteen compounds (1-14), including six alkaloids (1-3, 5, 7, and 8), two phenolic compounds (4 and 6), one fatty acid (9), and five steroids (10-14). The structures of the isolated compounds were determined using NMR spectroscopic methods, liquid chromatography-mass spectrometry, and comparison of data with previously reported values. The isolated compounds (1-14) were tested for cytotoxicity against MDA-MB-231 cells, where compound 1, i.e., N,N-dimethyl-anthranilic acid, exhibited the most significant cytotoxicity against MDA-MB-231 cells, with an IC50 value of 90.28 ± 4.23 μM and apoptotic cell death of 56.01% ± 2.64% at 100 μM. Treatment with compound 1 resulted in an upregulation of protein levels, including cleaved caspase-8, cleaved poly (ADP-ribose) polymerase, Bcl-2-associated X protein (Bax), cleaved caspase-3, cleaved caspase-9, Bad, and Cytochrome c, but decreased the levels of B-cell lymphoma 2 (Bcl-2). Overall, these results indicate that N,N-dimethyl-anthranilic acid (1) may have anti-breast cancer activity and is probably involved in the induction of apoptosis mediated by extrinsic and intrinsic signaling pathways.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Seulah Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Yoon Seo Jang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Rhim Ryoo
- Special Forest Products Division, Forest Bioresources Department, National Institute of Forest Science, Suwon 16631, Republic of Korea
| | - Jung Kyu Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
37
|
Li MY, Liu LZ, Xin Q, Zhou J, Zhang X, Zhang R, Wu Z, Yi J, Dong M. Downregulation of mTORC1 and Mcl-1 by lipid-oversupply contributes to islet β-cell apoptosis and dysfunction. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159332. [PMID: 37196823 DOI: 10.1016/j.bbalip.2023.159332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/22/2023] [Accepted: 04/30/2023] [Indexed: 05/19/2023]
Abstract
Pancreatic β-cell apoptosis is a key feature of diabetes and can be induced by chronic exposure to saturated fatty acids (FAs). However, the underlying mechanisms remain poorly understood. We presently evaluated the role of Mcl-1 and mTOR in mice fed with high-fat-diet (HFD) and β-cells exposed to the overloaded palmitic acid (PA). Compared with normal-chow-diet (NCD)-fed mice, HFD group showed impaired glucose tolerance after two months. Along with the diabetes progression, pancreatic islets first became hypertrophic and then atrophic, the ratio of β-cell:α-cell increased in the islets of four months HFD-fed mice while decreased after six months. This process was accompanied by significantly increased β-cell apoptosis and AMPK activity, and decreased Mcl-1 expression and mTOR activity. Consistently, glucose-induced insulin secretion dropped. In terms of mechanism, PA with lipotoxic dose could activate AMPK, which in turn inhibited ERK-stimulated Mcl-1Thr163 phosphorylation. Meanwhile, AMPK blocked Akt activity to release Akt inhibition on GSK3β, followed by GSK3β-initiated Mcl-1Ser159 phosphorylation. The context of Mcl-1 phosphorylation finally led to its degradation by ubiquitination. Also, AMPK inhibited the activity of mTORC1, resulting in a lower level of Mcl-1. Suppression of mTORC1 activity and Mcl-1 expression positively related to β-cell failure. Alteration of Mcl-1 or mTOR expression rendered different tolerance of β-cell to different dose of PA. In conclusion, lipid oversupply-induced dual modulation of mTORC1 and Mcl-1 finally led to β-cell apoptosis and impaired insulin secretion. The study may help further understand the pathogenesis of β-cell dysfunction in case of dyslipidemia, and provide promising therapeutic targets for diabetes.
Collapse
Affiliation(s)
- Ming-Yue Li
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China
| | - Li-Zhong Liu
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Qihang Xin
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Jiaying Zhou
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Xiaoyang Zhang
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Rui Zhang
- GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China
| | - Zangshu Wu
- GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China
| | - Junbo Yi
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Ming Dong
- GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China.
| |
Collapse
|
38
|
Masoudi R, Mohammadi A, Morovati S, Heidari AA, Asad-Sangabi M. Induction of apoptosis in colorectal cancer cells by matrix protein of PPR virus as a novel anti-cancer agent. Int J Biol Macromol 2023:125536. [PMID: 37369256 DOI: 10.1016/j.ijbiomac.2023.125536] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023]
Abstract
Colorectal cancer (CRC) is a common and highly malignant neoplasm, ranking as the fourth most frequent cause of cancer-related deaths worldwide. Recently, non-human oncolytic viruses such as Peste des petits ruminants virus (PPRV) are considered as a potent candidate in the viral therapy of cancer. In the current study, the apoptotic effects of matrix (M) protein of PPRV was investigated on SW480 CRC cells. The M gene was cloned into the pcDNA™3.1/Hygro(+) expression vector and transfected into the cancer cells. The cytotoxic effects of the M protein on SW480 cells were confirmed using MTT assay. Furthermore, flow cytometry results showed that the M protein induces apoptosis in 91 % of CRC cells. Interestingly, the expression of the M gene in SW480 cells led to the up-regulation of genes including Bax, p53, and Caspase-9, as well as an increase in the Bax/Bcl-2 ratio. By using bioinformatics modeling, we hypothesized that the M protein could interact with Bax factor through its BH3-like motif and could further activate the intrinsic apoptosis pathway. Ultimately, this study provided the first evidence of the pro-apoptotic activity of PPRV M protein indicating its possible development as a promising novel anti-cancer agent.
Collapse
Affiliation(s)
- Ramin Masoudi
- Division of Biotechnology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Mohammadi
- Division of Virology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Solmaz Morovati
- Division of Biotechnology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amir Ali Heidari
- Division of Aquatic Animal Health and Diseases, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mehdi Asad-Sangabi
- Division of Virology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
39
|
Li M, Yang G, Zheng Y, Lv J, Zhou W, Zhang H, You F, Wu C, Yang H, Liu Y. NIR/pH-triggered aptamer-functionalized DNA origami nanovehicle for imaging-guided chemo-phototherapy. J Nanobiotechnology 2023; 21:186. [PMID: 37301952 DOI: 10.1186/s12951-023-01953-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Targeted chemo-phototherapy has received widespread attention in cancer treatment for its advantages in reducing the side effects of chemotherapeutics and improving therapeutic effects. However, safe and efficient targeted-delivery of therapeutic agents remains a major obstacle. Herein, we successfully constructed an AS1411-functionalized triangle DNA origami (TOA) to codeliver chemotherapeutic drug (doxorubicin, DOX) and a photosensitizer (indocyanine green, ICG), denoted as TOADI (DOX/ICG-loaded TOA), for targeted synergistic chemo-phototherapy. In vitro studies show that AS1411 as an aptamer of nucleolin efficiently enhances the nanocarrier's endocytosis more than 3 times by tumor cells highly expressing nucleolin. Subsequently, TOADI controllably releases the DOX into the nucleus through the photothermal effect of ICG triggered by near-infrared (NIR) laser irradiation, and the acidic environment of lysosomes/endosomes facilitates the release. The downregulated Bcl-2 and upregulated Bax, Cyt c, and cleaved caspase-3 indicate that the synergistic chemo-phototherapeutic effect of TOADI induces the apoptosis of 4T1 cells, causing ~ 80% cell death. In 4T1 tumor-bearing mice, TOADI exhibits 2.5-fold targeted accumulation in tumor region than TODI without AS1411, and 4-fold higher than free ICG, demonstrating its excellent tumor targeting ability in vivo. With the synergetic treatment of DOX and ICG, TOADI shows a significant therapeutic effect of ~ 90% inhibition of tumor growth with negligible systemic toxicity. In addition, TOADI presents outstanding superiority in fluorescence and photothermal imaging. Taken together, this multifunctional DNA origami-based nanosystem with the advantages of specific tumor targeting and controllable drug release provides a new strategy for enhanced cancer therapy.
Collapse
Grants
- (12132004, U19A2006, 32171395) the National Natural Science Foundation of China
- (12132004, U19A2006, 32171395) the National Natural Science Foundation of China
- (23NSFSC0392, 23SYSX0108, 2022NSFSC0048) the Sichuan Science and Technology Program
- (23NSFSC0392, 23SYSX0108, 2022NSFSC0048) the Sichuan Science and Technology Program
- (ZYGX2021YGLH204, ZYGX2021YGLH017, ZYGX2021YGLH023) the Joint Funds of Center for Engineering Medicine
- (ZYGX2021YGLH204, ZYGX2021YGLH017, ZYGX2021YGLH023) the Joint Funds of Center for Engineering Medicine
Collapse
Affiliation(s)
- Mengyue Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Geng Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Yue Zheng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Jiazhen Lv
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Wanyi Zhou
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Hanxi Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan, 610072, P.R. China
| | - Chunhui Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Hong Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China.
| | - Yiyao Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan, 610072, P.R. China.
| |
Collapse
|
40
|
Alshehri KM, Abdella EM. Development of ternary nanoformulation comprising bee pollen-thymol oil extracts and chitosan nanoparticles for anti-inflammatory and anticancer applications. Int J Biol Macromol 2023; 242:124584. [PMID: 37100316 DOI: 10.1016/j.ijbiomac.2023.124584] [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: 01/08/2023] [Revised: 04/05/2023] [Accepted: 04/20/2023] [Indexed: 04/28/2023]
Abstract
Due to the beneficial nutritional and medicinal characteristics of bee honey and thymol oil as antioxidants, anti-inflammatory agents, and antibacterial agents, they have been used since ancient times. The current study aimed to construct a ternary nanoformulation (BPE-TOE-CSNPs NF) through the immobilization of the ethanolic extract of bee pollen (BPE) with thymol oil extract (TOE) into the matrix of chitosan nanoparticles (CSNPs). The antiproliferative activity of new NF (BPE-TOE-CSNPs) against HepG2 and MCF-7 cells was investigated. The BPE-TOE-CSNPs showed significant inhibitory activity for the production of the inflammatory cytokines in HepG2 and MCF-7, with p < 0.001 for both TNF-α and IL6. Moreover, the encapsulation of the BPE and TOE in CSNPs increased the efficacy of the treatment and the induction of valuable arrests for the S phase of the cell cycle. In addition, the new NF has a great capacity to trigger apoptotic mechanisms through caspase-3 expression upregulation in cancer cells by two-fold among HepG2 cell lines and nine-fold among MCF-7 which appeared to be more susceptible to the nanoformulation. Moreover, the nanoformulated compound has upregulated the expression of caspase-9 and P53 apoptotic mechanisms. This NF may shed light on its pharmacological actions by blocking specific proliferative proteins, inducing apoptosis, and interfering with the DNA replication process.
Collapse
Affiliation(s)
- Kulud M Alshehri
- Department of Biology, Al Baha University, Baljurashi, Saudi Arabia.
| | - Ehab M Abdella
- Department of Biology, Al Baha University, Al Aqiq, Saudi Arabia; Department of Zoology, Faculty of Science, Beni Suaif University, Egypt
| |
Collapse
|
41
|
Singh S, Rani H, Sharma N, Behl T, Zahoor I, Makeen HA, Albratty M, Alhazm HA, Aleya L. Targeting multifunctional magnetic nanowires for drug delivery in cancer cell death: an emerging paradigm. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:57219-57235. [PMID: 37010687 DOI: 10.1007/s11356-023-26650-w] [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: 12/16/2021] [Accepted: 03/21/2023] [Indexed: 05/10/2023]
Abstract
Apoptosis, often known as programmed cell death is a mechanism used by numerous species to maintain tissue homeostasis. The process leading to cell death is complicated because it requires the stimulation of caspases. According to several studies, nanowires have important medical benefits, can kill cells by adhering to cancer cells, destroying them, and killing the entire cell using a triple attack that integrates vibration, heat, and drug delivery to trigger apoptosis. The sewage effluents and industrial, fertilizer and organic wastes decomposition can produce elevated levels of chemicals in the environment which may interrupt the cell cycle and activate apoptosis. The purpose of this review is to give a thorough summary of the evidence that is currently available on apoptosis. Current review discussed topics like the morphological and biochemical alterations that occur during apoptosis, as well as the various mechanisms that cause cell death, including the intrinsic (or mitochondrial), extrinsic (or death receptor), and intrinsic endoplasmic reticulum pathway. The apoptosis reduction in cancer development is mediated by (i) an imbalance between pro- and anti-apoptotic proteins, such as members of the B-cell lymphoma-2 (BCL2) family of proteins, tumour protein 53 and inhibitor of apoptosis proteins, (ii) a reduction in caspase activity, and (iii) impaired death receptor signalling. This review does an excellent task of outlining the function of nanowires in both apoptosis induction and targeted drug delivery for cancer cells. A comprehensive summary of the relevance of nanowires synthesised for the purpose of inducing apoptosis in cancer cells has been compiled collectively.
Collapse
Affiliation(s)
- Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Hema Rani
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana, 141104, India
| | - Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India.
| | - Tapan Behl
- School of Health Sciences &Technology, University of Petroleum and Energy Studies, Bidholi, Uttarakhand, 248007, Dehradun, India
| | - Ishrat Zahoor
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A Alhazm
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| |
Collapse
|
42
|
Targeting mitochondria as a potential therapeutic strategy against chemoresistance in cancer. Biomed Pharmacother 2023; 160:114398. [PMID: 36773523 DOI: 10.1016/j.biopha.2023.114398] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/21/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023] Open
Abstract
The importance of mitochondria is not only limited to energy generation but also in several physical and chemical processes critical for cell survival. Mitochondria play an essential role in cellular apoptosis, calcium ion transport and cellular metabolism. Mutation in the nuclear and mitochondrial genes, altered oncogenes/tumor suppressor genes, and deregulated signalling for cell viability are major reasons for cancer progression and chemoresistance. The development of drug resistance in cancer patients is a major challenge in cancer treatment as the resistant cells are often more aggressive. The drug resistant cells of numerous cancer types exhibit the deregulation of mitochondrial function. The increased biogenesis of mitochondria and its dynamic alteration contribute to developing resistance. Further, a small subpopulation of cancer stem cells in the heterogeneous tumor is primarily responsible for chemoresistance and has an attribute of mitochondrial dysfunction. This review highlights the critical role of mitochondrial dysfunction in chemoresistance in cancer cells through the processes of apoptosis, autophagy/mitophagy, and cancer stemness. Mitochondria-targeted therapeutic strategies might help reduce cancer progression and chemoresistance induced by various cancer drugs.
Collapse
|
43
|
Tavakoli Pirzaman A, Aghajanian S, Mansoori R, Al-E-Ahmad A, Ebrahimzadeh M, Moghadamnia AA, Kazemi S. Interaction of quercetin and 5-fluorouracil: cellular and pharmacokinetic study. Toxicol Mech Methods 2023:1-10. [PMID: 36912048 DOI: 10.1080/15376516.2023.2188928] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
5-fluorouracil (5-FU) is a widely used chemotherapeutic agent, and its uncontrolled blood levels contribute to toxicity. Quercetin, as an important flavonoid, has many biological effects, including anti-tumor and anti-inflammatory features. The current study investigated the synergistic effect between 5-FU and quercetin using HT-29 cell line and fibroblast cells. Rats were assigned to two groups. The 5-FU/quercetin group received intraperitoneal quercetin (10 mg/kg) and the Tween was injected to the control group for 14 consecutive days. On the 15th day, both groups received 50 mg/kg of 5-FU. Upon the final injection, blood samples were obtained at different times. Pharmacokinetic parameters were evaluated using high-performance liquid chromatography (HPLC). The mean (±SD) of maximum plasma concentration (Cmax) of 5-FU in combination therapy group was 3.10 ± 0.18 μg/ml and the area under the curve (AUC) was 153.89 ± 21.36, which increased by 113% and 128% compared to control group, respectively. Quercetin increased anti-tumor activity of 5-FU and enhanced Cmax and AUC of 5-FU. These findings confirm the synergistic effects between quercetin and 5-FU at the usual doses in cancer treatment, which may lead to reduced toxicity.
Collapse
Affiliation(s)
- Ali Tavakoli Pirzaman
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Soheyla Aghajanian
- Department of Pharmacology, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | - Razieh Mansoori
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Atiyeh Al-E-Ahmad
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Manijeh Ebrahimzadeh
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
44
|
Yang HL, Huang ST, Lyu ZH, Bhat AA, Vadivalagan C, Yeh YL, Hseu YC. The anti-tumor activities of coenzyme Q0 through ROS-mediated autophagic cell death in human triple-negative breast cells. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
|
45
|
Genetic mutations affecting mitochondrial function in cancer drug resistance. Genes Genomics 2023; 45:261-270. [PMID: 36609747 PMCID: PMC9947062 DOI: 10.1007/s13258-022-01359-1] [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: 11/15/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023]
Abstract
Mitochondria are organelles that serve as a central hub for physiological processes in eukaryotes, including production of ATP, regulation of calcium dependent signaling, generation of ROS, and regulation of apoptosis. Cancer cells undergo metabolic reprogramming in an effort to support their increasing requirements for cell survival, growth, and proliferation, and mitochondria have primary roles in these processes. Because of their central function in survival of cancer cells and drug resistance, mitochondria are an important target in cancer therapy and many drugs targeting mitochondria that target the TCA cycle, apoptosis, metabolic pathway, and generation of ROS have been developed. Continued use of mitochondrial-targeting drugs can lead to resistance due to development of new somatic mutations. Use of drugs is limited due to these mutations, which have been detected in mitochondrial proteins. In this review, we will focus on genetic mutations in mitochondrial target proteins and their function in induction of drug-resistance.
Collapse
|
46
|
PW06 Triggered Fas-FADD to Induce Apoptotic Cell Death In Human Pancreatic Carcinoma MIA PaCa-2 Cells through the Activation of the Caspase-Mediated Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3479688. [PMID: 36820406 PMCID: PMC9938777 DOI: 10.1155/2023/3479688] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/25/2022] [Accepted: 09/21/2022] [Indexed: 02/13/2023]
Abstract
Pancreatic cancer has higher incidence and mortality rates worldwide. PW06 [(E)-3-(9-ethyl-9H-carbazol-3-yl)-1-(2,5-dimethoxyphenyl) prop-2-en-1-one] is a carbazole derivative containing chalcone moiety which was designed for inhibiting tumorigenesis in human pancreatic cancer. This study is aimed at investigating PW06-induced anticancer effects in human pancreatic cancer MIA PaCa-2 cells in vitro. The results showed PW06 potent antiproliferative/cytotoxic activities and induced cell morphological changes in a human pancreatic cancer cell line (MIA PaCa-2), and these effects are concentration-dependent (IC50 is 0.43 μM). Annexin V and DAPI staining assays indicated that PW06 induced apoptotic cell death and DNA condensation. Western blotting indicated that PW06 increased the proapoptotic proteins such as Bak and Bad but decreased the antiapoptotic protein such as Bcl-2 and Bcl-xL. Moreover, PW06 increased the active form of caspase-8, caspase-9, and caspase-3, PARP, releasing cytochrome c, AIF, and Endo G from mitochondria in MIA PaCa-2 cells. Confocal laser microscopy assay also confirmed that PW06 increased Bak and decreased Bcl-xL. Also, the cells were pretreated with inhibitors of caspase-3, caspase-8, and caspase-9 and then were treated with PW06, resulting in increased viable cell number compared to PW06 treated only. Furthermore, PW06 showed a potent binding ability with hydrophobic interactions in the core site of the Fas-Fas death domains (FADD). In conclusion, PW06 can potent binding ability to the Fas-FADD which led to antiproliferative, cytotoxic activities, and apoptosis induction accompanied by the caspase-dependent and mitochondria-dependent pathways in human pancreatic cancer MIA PaCa-2 cells.
Collapse
|
47
|
Guo XJ, Huang XY, Yang X, Lu JC, Wei CY, Gao C, Pei YZ, Chen Y, Sun QM, Cai JB, Zhou J, Fan J, Ke AW, Shi YG, Shen YH, Zhang PF, Shi GM, Yang GH. Loss of 5-hydroxymethylcytosine induces chemotherapy resistance in hepatocellular carcinoma via the 5-hmC/PCAF/AKT axis. Cell Death Dis 2023; 14:79. [PMID: 36732324 PMCID: PMC9895048 DOI: 10.1038/s41419-022-05406-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 02/04/2023]
Abstract
Multidrug resistance is a major challenge in treating advanced hepatocellular carcinoma (HCC). Although recent studies have reported that the multidrug resistance phenotype is associated with abnormal DNA methylation in cancer cells, the epigenetic mechanism underlying multidrug resistance remains unknown. Here, we reported that the level of 5-hydroxymethylcytosine (5-hmC) in human HCC tissues was significantly lower than that in adjacent liver tissues, and reduced 5-hmC significantly correlated with malignant phenotypes, including poor differentiation and microvascular invasion; additionally, loss of 5-hmC was related to chemotherapy resistance in post-transplantation HCC patients. Further, the 5-hmC level was regulated by ten-eleven translocation 2 (TET2), and the reduction of TET2 in HCC contributes to chemotherapy resistance through histone acetyltransferase P300/CBP-associated factor (PCAF) inhibition and AKT signaling hyperactivation. In conclusion, loss of 5-hmC induces chemotherapy resistance through PCAF/AKT axis and is a promising chemosensitivity prediction biomarker and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Xiao-Jun Guo
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Xiao-Yong Huang
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Xuan Yang
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
- Department of General Surgery, Peking University Third Hospital, Beijing, PR China
| | - Jia-Cheng Lu
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Chuan-Yuan Wei
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Chao Gao
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Yan-Zi Pei
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Yi Chen
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Qi-Man Sun
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
| | - Jia-Bin Cai
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
| | - Jian Zhou
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Jia Fan
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Ai-Wu Ke
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China
| | - Yujiang G Shi
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, PR China.
| | - Ying-Hao Shen
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China.
| | - Peng-Fei Zhang
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China.
- Department of Medical Oncology, Zhongshan Hospital of Fudan University, Shanghai, 200032, PR China.
- Cancer Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, PR China.
| | - Guo-Ming Shi
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China.
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China.
- Clinical Research Unit, Institute of Clinical Science, Zhongshan Hospital of Fudan University, Shanghai, 200032, PR China.
| | - Guo-Huan Yang
- Department of Liver Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital of Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China.
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, 200032, PR China.
| |
Collapse
|
48
|
Takaya K, Ishii T, Asou T, Kishi K. Navitoclax (ABT-263) Rejuvenates Human Skin by Eliminating Senescent Dermal Fibroblasts in a Mouse/Human Chimeric Model. Rejuvenation Res 2023; 26:9-20. [PMID: 36324221 DOI: 10.1089/rej.2022.0048] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Chronic senescence, such as aging, contributes to age-related tissue dysfunction and disease development. The accumulation of senescent fibroblasts and the senescence-associated secretory phenotype is particularly implicated in this process. Removal of senescent cells has been reported to prevent tissue dysfunction and to extend the life span during aging. ABT-263 (navitoclax), which inhibits antiapoptotic proteins, is a leading antiaging drug; however, its role in human skin aging is unclear. This study aimed to determine the rejuvenating effects of ABT-263 on aging skin using a human skin graft mouse model. We assessed the viability of ABT-263-treated skin fibroblasts after inducing senescence. Aged human skin was transplanted under the back skin of nude mice and injected intraperitoneally with the drug or control. Analysis of the skin specimens revealed that ABT-263 induced selective elimination of senescent dermal fibroblasts. Senescent human skin treated with ABT-263 exhibited a decrease in the number of senescent cells and in the expression of aging-related secretory phenotype molecules, such as matrix metalloproteinases and interleukins and an increase in collagen density. Our results indicate that selective removal of senescent skin cells with ABT-263 can improve the aging phenotype of human skin without side effects. ABT-263 is, thus, a novel potential therapeutic agent for skin aging.
Collapse
Affiliation(s)
- Kento Takaya
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuyuki Ishii
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Toru Asou
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
Su W, Hu Y, Fan X, Xie J. Clearance of senescent cells by navitoclax (ABT263) rejuvenates UHMWPE-induced osteolysis. Int Immunopharmacol 2023; 115:109694. [PMID: 36638657 DOI: 10.1016/j.intimp.2023.109694] [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: 09/28/2022] [Revised: 12/13/2022] [Accepted: 01/02/2023] [Indexed: 01/13/2023]
Abstract
Periprosthetic osteolysis is the leading cause of prosthesis failure and subsequent total joint revision. Wear particles produced by prosthetic materials are the main biological factors that cause periprosthetic osteolysis. Reducing the inflammatory response induced by the phagocytosis of wear particles by macrophages, blocking the activation of osteoclastogenesis, and promoting bone regeneration are essential for preventing the aseptic loosening of prostheses. In this study, we demonstrated that cellular senescence played a vital role during the process of ultra-high molecular weight polyethylene (UHMWPE) particle-induced osteolysis. Administration of the senolytic drug navitoclax (ABT263) could eliminate senescent cells and inhibit the secretion and inflammatory state of the senescence-associated secretory phenotype (SASP). We also discovered that ABT263 inhibited the formation of osteoclasts and had a significant therapeutic effect on UHMWPE particle-induced osteolysis based on the results of UHMWPE-induced mouse cranial osteolysis. Therefore, our research provided innovative strategies and ideas for the prevention and treatment of periprosthetic osteolysis.
Collapse
Affiliation(s)
- Weiping Su
- Department of Orthopedics, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolei Fan
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.
| | - Jie Xie
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
50
|
Effect of Formaldehyde and Curcumin on Histomorphological Indices, Gene Expression Associated with Ovarian Follicular Development, and Total Antioxidant to Oxidant Levels in Wistar Rats. Int J Biomater 2023; 2023:4662440. [PMID: 36776151 PMCID: PMC9908331 DOI: 10.1155/2023/4662440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Abstract
The present experimental study was undertaken to investigate the effect of formaldehyde (FA) and curcumin (CUR) on histomorphological features, antioxidant potential, and messenger ribonucleic acid (mRNA) levels of genes related to follicular development in FA-exposed rats. 24 Wistar female rats were divided into four study groups and given intraperitoneal injections of FA (10 mg/kg) (N = 6), FA (10 mg/kg) + CUR (100 mg/kg) (N = 6), sham (N = 6), and control (N = 6) for 14 days. Ovarian follicular histology, the related gene expression, blood factors, and anti/oxidation potentials were assessed using ovarian tissue and serum, respectively. The klotho was significantly overexpressed in the FA group compared with controls and shams. Contradictory, the factor in germ line alpha was significantly down-regulated in FA and FA + CUR groups compared to shams and controls. A significant decline was seen in the number of ovarian follicles in the FA group, independent of the developmental stage. Regarding the comparison of the FA + CUR group to other groups, a significant change was seen in the number of secondary, graafian, and atretic follicles. The FA group demonstrated significantly lower hemoglobin, red blood cell count, hematocrit, and mean corpuscular hemoglobin concentration than controls. The activity of glutathione peroxidase increased significantly in the FA group than in the controls. Despite the deleterious effects of FA on histological and molecular aspects of rat ovarian follicles, CUR does not appear to have a protective effect against the hazardous effects of this chemical. However, CUR in some cases has positive effects such as reducing follicular destruction and interstitial edema.
Collapse
|