1
|
Gomes I, Oliveira RJDS, Girol AP. Signaling pathways in glioblastoma. Crit Rev Oncol Hematol 2025; 209:104647. [PMID: 39961403 DOI: 10.1016/j.critrevonc.2025.104647] [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: 11/10/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Cancer is one of the main public health problems worldwide. Among tumors of the Central Nervous System (CNS), glioblastoma (GBM) affects 49.1 % of malignant brain tumors, and despite standard treatment, patients diagnosed with GBM have a dismal prognosis, a high rate of recurrence after tumor resection and poor survival. Since 2016, the World Health Organization (WHO) has included molecular biomarkers in the classification of these tumors, as knowing the heterogeneity and possible genetic changes allows for new therapeutic possibilities. The purpose of this review was to provide an overview of epidemiology and classification, as well as changes in signaling pathways resulting from genetic alterations that affect crucial factors in tumorigenesis, response to treatment and prognosis. Therefore, understanding and characterizing the vast genetic heterogeneity of GBM, both genetic and epigenetic alterations, enable a greater comprehension of the pathogenesis of this tumor, potentially helping to bring new therapeutic approaches and personalization of treatment through the different genetic alterations in each patient.
Collapse
Affiliation(s)
- Isabella Gomes
- Department of Biology, Post Graduate Program in Biosciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, Brazil; Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva, SP, Brazil
| | | | - Ana Paula Girol
- Department of Biology, Post Graduate Program in Biosciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, Brazil; Experimental and Clinical Research Center (CEPEC), Padre Albino University Center (UNIFIPA), Catanduva, SP, Brazil; Post Graduate Program in Structural and Functional Biology, Paulista School of Medicine (UNIFESP-EPM), Federal University of São Paulo, SP, Brazil.
| |
Collapse
|
2
|
Alamillo-Vásquez JA, Pérez-Torres CA, Ibarra-Laclette E, Ramón-Farías F, Nicasio-Torres P, Alatorre-Cobos F. Secondary Metabolites from Croton Species and Their Biological Activity on Cell Cycle Regulators. Metabolites 2025; 15:216. [PMID: 40278345 PMCID: PMC12029420 DOI: 10.3390/metabo15040216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Plant-based traditional medicine integrates beliefs, knowledge, and practices to prevent and treat multiple diseases. Croton is a large and worldwide-spread genus belonging to Euphorbiaceae, a family well known for comprising many species with medicinal properties due to its high diversity of phytochemical constituents with biological activities. Among the various benefits of Croton species in traditional medicine, its use in cancer treatment has recently received significant attention from the scientific community. This review provides a general overview of different studies on the Croton genus in the research for alternative cancer treatments and the impact of its secondary metabolite catalog on cell cycle targets. Our analysis indicates that just under 30 secondary metabolites have been identified so far in latex and extracts obtained from leaves, twigs, or bark from 22 different Croton species. Based on standard assays using cell lines or human platelets, these molecules show multiple biological activities mainly compromising cell viability and cell cycle progression, supporting the ethnobotanical use of Croton species for cancer treatment. Several studies indicate that Croton metabolites target CDK-cyclin complexes and signaling routes that trigger apoptosis; however, further studies are needed to better understand the molecular mechanisms underlying Croton metabolites' effects and their accurate future applications in cancer treatment.
Collapse
Affiliation(s)
- Jorge Augusto Alamillo-Vásquez
- Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico; (J.A.A.-V.); (E.I.-L.)
| | - Claudia-Anahí Pérez-Torres
- SECIHTI-Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico
| | - Enrique Ibarra-Laclette
- Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico; (J.A.A.-V.); (E.I.-L.)
| | - Feliza Ramón-Farías
- Facultad de Ciencias Biológicas y Agropecuarias, Universidad Veracruzana, Córdoba 94500, Veracruz, Mexico;
| | - Pilar Nicasio-Torres
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62790, Morelos, Mexico;
| | - Fulgencio Alatorre-Cobos
- SECIHTI-Unidad de Biología Integrativa Centro de Investigación Científica de Yucatán, A.C., Calle 43 No. 130 × 32 y 34, Col. Chuburná de Hidalgo, Mérida 97205, Yucatán, Mexico
| |
Collapse
|
3
|
Casso-Chapa B, González NAV, Le NT, Palaskas NL, Nead KT, Eutsey LP, Samanthapudi VSK, Osborn AM, Lee J, Mejia G, Hoang O, Lin SH, Deswal A, Herrmann J, Wang G, Kirkland JL, Krishnan S, Wehrens XH, Chini EN, Yusuf SW, Iliescu CA, Jain A, Burks JK, Seeley E, Lorenzi PL, Chau KM, Mendoza KCO, Grumbach IM, Brookes PS, Hanssen NM, de Winther MP, Yvan-Charvet L, Kotla S, Schadler K, Abe JI. Reevaluating Anti-Inflammatory Therapy: Targeting Senescence to Balance Anti-Cancer Efficacy and Vascular Disease. Arterioscler Thromb Vasc Biol 2025; 45:372-385. [PMID: 39817327 PMCID: PMC11864897 DOI: 10.1161/atvbaha.124.319870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025]
Abstract
Modulating immune function is a critical strategy in cancer and atherosclerosis treatments. For cancer, boosting or maintaining the immune system is crucial to prevent tumor growth. However, in vascular disease, mitigating immune responses can decrease inflammation and slow atherosclerosis progression. Anti-inflammatory therapy, therefore, presents a unique dilemma for cancer survivors: while it may decrease cardiovascular risk, it might also promote cancer growth and metastasis by suppressing the immune response. Senescence presents a potentially targetable solution to this challenge; senescence increases the risk of both cancer therapy resistance and vascular disease. Exercise, notably, shows promise in delaying this premature senescence, potentially improving cancer outcomes and lowering vascular disease risk post-treatment. This review focuses on the long-term impact of cancer therapies on vascular health. We underscore the importance of modulating senescence to balance cancer treatment's effectiveness and its vascular impact, and we emphasize investigating the role of exercise-mediated suppression of senescence in improving cancer survivorship.
Collapse
Affiliation(s)
- Bernardo Casso-Chapa
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Norma Alicia Vazquez González
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kevin T. Nead
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lydia P. Eutsey
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Cancer Center Support Grant & Extramural Research Development, UT MD Anderson Cancer Center, Houston, TX
| | | | - Abigail M Osborn
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonghae Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gilbert Mejia
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Oanh Hoang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xander H.T. Wehrens
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cezar A. Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jared K. Burks
- Department of Leukemia, Division of Center Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Erin Seeley
- Department of Chemistry, University of Texas at Austin, Austin, Texas, USA
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Khanh M. Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Keila Carolina Ostos Mendoza
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | | | - Paul S. Brookes
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Nordin M.J. Hanssen
- Department of (Experimental) Vascular and Internal Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Diabeter Centrum Amsterdam, Amsterdam, the Netherlands
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Immunology and Infectious Diseases (AII), Inflammatory Diseases Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keri Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Ahmed S, Aschner M, Alsharif KF, Allahyani M, Huang G, Wan C, Khan H. Marine peptides in lymphoma: surgery at molecular level for therapeutic understanding. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03901-w. [PMID: 39992419 DOI: 10.1007/s00210-025-03901-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Lymphoma, the most common form of blood cancer, affects primarily the intricate network of tissues and organs known as the lymphatic system. Globally, it ranks among the leading causes of cancer-related deaths. Although conventional therapies have led to significant advancements, they are accompanied by adverse side effects and present challenges in cases of multidrug resistance, refractory patients, and relapses. This highlights a pressing need for innovative treatment approaches. Extensive research on the anti-lymphoma properties of natural compounds has particularly focused on marine organisms as valuable sources for potential medicinal agents. Among these, anticancer peptides have garnered attention due to their multiple beneficial effects against cancer, coupled with reduced toxicity to normal cells. This review focuses on the molecular mechanisms underlying the anti-lymphoma effects of marine peptides, examining the diverse pathways through which these peptides impact physiological processes. Key effects include modulation of cell viability, induction of apoptosis, cell cycle arrest, antimitotic activity, immunotherapeutic properties, disruption of mitochondrial function and induction of oxidative stress, cancer cell membrane destruction, and interference with microtubule stability. The review also highlights the antibody-drug conjugates (ADCs) derived from marine peptides and their synergistic effects with other anti-lymphoma medications. This knowledge should inspire future study and development of these prospective therapeutic modalities and hasten the investigation and creation of novel lymphoma remedies derived from marine sources.
Collapse
Affiliation(s)
- Salman Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Khalaf F Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Mamdouh Allahyani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Guang Huang
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chunpeng Wan
- Jiangxi Provincial Key Laboratory for Postharvest Storage and Preservation of Fruits & Vegetables, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa, 23200, Pakistan.
- Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| |
Collapse
|
5
|
Shahrokhi H, Asili J, Tayarani-Najaran Z, Boozari M. Signaling pathways behind the biological effects of tanshinone IIA for the prevention of cancer and cardiovascular diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03857-x. [PMID: 39937254 DOI: 10.1007/s00210-025-03857-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Tanshinone IIA (Tan IIA) is a well-known fat-soluble diterpenoid found in Salvia miltiorrhiza, recognized for its various biological effects. The molecular signaling pathways of Tan IIA have been investigated in different diseases, including the anti-inflammatory, hepatoprotective, renoprotective, neuroprotective effects, and fibrosis prevention. This article provides a brief overview of the signaling pathways related to anti-cancer and cardioprotective effects of Tan IIA. It shows that Tan IIAs anti-cancer ability has good expectation through multiplicity mechanisms affecting various aspects' tumor biology. The major pathways involved in its anti-cancer effects include inhibition of PI3/Akt, MAPK, and p53/p21 signaling which leads to enhancement of immune responses and increased radiation sensitivity. Some essential pathways responsible for cardioprotective effects induced by Tan IIA are PI3/AKT activation, MAPK, and SIRT1 promoting protection against ischemia/reperfusion injury in myocardial cells as well as inhibiting pathological remodeling processes. Finally, the article underscores the complex and specific signaling pathways influenced by Tan IIA. The PI3/Akt and MAPK pathways play critical roles in the anti-cancer and cardioprotective effects of Tan IIA. Particularly, Tan IIA suppresses the proliferation of malignancies in cancerous cells but stimulates protective mechanisms in normal cardiovascular cells. These findings highlight the importance of investigating molecular signaling pathways in evaluating the therapeutic potential of natural products. Studying about signaling pathways is vital in understanding the therapeutic aspects of Tan IIA and its derivatives as anti-cancer and cardio-protective agents. Further research is necessary to understand these complex mechanisms.
Collapse
Affiliation(s)
- Homa Shahrokhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Motahareh Boozari
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Brane A, Sutko M, Tollefsbol TO. p21 Promoter Methylation Is Vital for the Anticancer Activity of Withaferin A. Int J Mol Sci 2025; 26:1210. [PMID: 39940977 PMCID: PMC11818515 DOI: 10.3390/ijms26031210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Breast cancer (BC) is a widespread malignancy that affects the lives of millions of women each year, and its resulting financial and healthcare hardships cannot be overstated. These issues, in combination with side effects and obstacles associated with the current standard of care, generate considerable interest in new potential targets for treatment as well as means for BC prevention. One potential preventive compound is Withaferin A (WFA), a traditional medicinal compound found in winter cherries. WFA has shown promise as an anticancer agent and is thought to act primarily through its effects on the epigenome, including, in particular, the methylome. However, the relative importance of specific genes' methylation states to WFA function remains unclear. To address this, we utilized human BC cell lines in combination with CRISPR-dCas9 fused to DNA methylation modifiers (i.e., epigenetic editors) to elucidate the importance of specific genes' promoter methylation states to WFA function and cancer cell viability. We found that targeted demethylation of promoters of the tumor suppressors p21 and p53 within MDA-MB-231/MCF7 cells resulted in around 1.7×/1.5× and 1.2×/1.3× increases in expression, respectively. Targeted methylation of the promoter of the oncogene CCND1 within MDA-MB-231/MCF7 cells resulted in 0.5×/0.8× decreases in gene expression. These changes to p21, p53, and CCND1 were also associated with decreases in cell viability of around 25%/50%, 5%/35%, and 12%/16%, respectively, for MDA-MB-231/MCF7 cells. When given in combination with WFA in both p53 mutant and wild type cells, we discovered that targeted methylation of the p21 promoter was able to modulate the anticancer effects of WFA, while targeted methylation or demethylation of the promoters of p53 and CCND1 had no significant effect on viability decreases from WFA treatment. Taken together, these results indicate that p21, p53, and CCND1 may be important targets for future in vivo studies that may lead to epigenetic editing therapies and that WFA may have utility in the prevention of BC through its effect on p21 promoter methylation independent of p53 function.
Collapse
Affiliation(s)
- Andrew Brane
- Department of Biology, University of Alabama at Birmingham, 3100 Science & Engineering Complex—East Science Hall, 902 14th Street South, Birmingham, AL 35205, USA
| | - Madeline Sutko
- Department of Biology, University of Alabama at Birmingham, 3100 Science & Engineering Complex—East Science Hall, 902 14th Street South, Birmingham, AL 35205, USA
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 3100 Science & Engineering Complex—East Science Hall, 902 14th Street South, Birmingham, AL 35205, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA
- Integrative Center for Aging Research, University of Alabama at Birmingham, 933 19th Street South, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, 1675 University Blvd, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Yu Y, Zhu C, Wang X, Shi Y, Gao Y, Yu Z. hERG activators exhibit antitumor effects in breast cancer through calcineurin and β-catenin-mediated signaling pathways. Front Pharmacol 2025; 16:1545300. [PMID: 39917621 PMCID: PMC11799564 DOI: 10.3389/fphar.2025.1545300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Background Breast cancer remains a leading cause of mortality among women worldwide, with existing therapeutic options often accompanied by significant side effects and a persistent risk of disease recurrence. This highlights the need for novel drug candidates with new mechanisms of action by targeting alternative signaling pathways. While hERG channel is notoriously regarded as an off-target due to drug-induced cardiotoxicity, its therapeutic potential as a drug target remains largely unexplored. Methods This study investigated the role of hERG in breast cancer progression and its impact on patient survival. The anti-proliferative, anti-migratory, anti-invasive and pro-apoptotic effects of hERG activators were evaluated using the Cell Counting Kit-8, wound healing assay, transwell assay and cell apoptosis assay, respectively. Western blotting, Ca2+ imaging and immunofluorescence assays were employed to study their antitumor mechanisms of actions. Results We identified two novel hERG activators, SDUY429 and SDUY436, which effectively inhibited the proliferation and migration of MDA-MB-231 and MCF-7 cells. In addition, SDUY436 demonstrated significant anti-invasive and pro-apoptotic effects in MDA-MB-231 cells. Mechanistically, the anti-proliferative activity of hERG activators were mediated through calcineurin activation via enhanced calcium ion influx, which facilitated the nuclear translocation of nuclear factor of activated T cells (NFAT) and upregulated p21Waf/Cip expression. Furthermore, both SDUY429 and SDUY436 remarkably suppressed the migration and invasion of MDA-MB-231 cells by downregulating the protein kinase B (AKT)/glycogen synthase kinase-3 beta (GSK3β)/β-catenin signaling pathway. The observed reduction in phospho-AKT-Ser473 (pAKTS473) expression resulted in the decreased levels of phospho-GSK3β-Ser9 (pGSK3βS9), thereby limiting the nuclear localization of β-catenin, which led to the inhibition of cell migration and invasion. Notably, combining SDUY429 or SDUY436 with the AKT inhibitor MK-2206 produced synergistic anti-proliferative effects. Conclusion These findings suggest that hERG activators hold promise as new potential therapeutic agents for the treatment of breast cancer, paving the way for future investigations into their clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhiyi Yu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
8
|
Gupta S, Silveira DA, Mombach JCM, Hashimoto RF. DNA Damage-Induced Ferroptosis: A Boolean Model Regulating p53 and Non-Coding RNAs in Drug Resistance. Proteomes 2025; 13:6. [PMID: 39846637 PMCID: PMC11755436 DOI: 10.3390/proteomes13010006] [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: 11/14/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
The tumor suppressor p53, in its wild-type form, plays a central role in cellular homeostasis by regulating senescence, apoptosis, and autophagy within the DNA damage response (DDR). Recent findings suggest that wild-type p53 also governs ferroptosis, an iron-dependent cell death process driven by lipid peroxidation. Post-translational modifications of p53 generate proteoforms that significantly enhance its functional diversity in regulating these mechanisms. A key target in this process is the cystine/glutamate transporter (xCT), which is essential for redox balance and ferroptosis resistance. Additionally, p53-induced miR-34c-5p suppresses cancer cell proliferation and drug resistance by modulating Myc, an oncogene further influenced by non-coding RNAs like circular RNA NOTCH1 (CricNOTCH1) and long non-coding RNA MALAT1. However, the exact role of these molecules in ferroptosis remains unclear. To address this, we introduce the first dynamic Boolean model that delineates the influence of these ncRNAs and p53 on ferroptosis, apoptosis, and senescence within the DDR context. Validated through gain- and loss-of-function perturbations, our model closely aligns with experimental observations in cancers such as oral squamous cell carcinoma, nasopharyngeal carcinoma, and osteosarcoma. The model identifies crucial positive feedback loops (CricNOTCH1/miR-34c/Myc, MALAT1/miR-34c/Myc, and Myc/xCT) and highlights the therapeutic potential of using p53 proteoforms and ncRNAs to combat drug resistance and induce cancer cell death.
Collapse
Affiliation(s)
- Shantanu Gupta
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, São Paulo 05508-090, SP, Brazil;
| | | | - José Carlos M. Mombach
- Departamento de Física, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil;
| | - Ronaldo F. Hashimoto
- Instituto de Matemática e Estatística, Departamento de Ciência da Computação, Universidade de São Paulo, Rua do Matão 1010, São Paulo 05508-090, SP, Brazil;
| |
Collapse
|
9
|
Arun D, Rath SL. Structural analysis of the impact of germline mutations of p16 in melanoma prone families. Mol Divers 2025:10.1007/s11030-024-11089-z. [PMID: 39821174 DOI: 10.1007/s11030-024-11089-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Cyclin-dependent kinases (CDKs), play essential roles in cell cycle progression. CDK activity is controlled through phosphorylation and inhibition by CDK inhibitors, such as p16. Mutations in p16 can lead to diseases such as cancer. This study examines a series of p16 mutants and their molecular interactions with CDK4 using modelling, molecular dynamics simulations, and docking studies. Despite no significant structural changes in p16 due to mutation, the binding affinity was found to be affected, correlating with conservation scales. Simulations revealed that specific mutations, such as G23D, P114S, and A60V resulted in loss of binding to CDK4, while others like R24Q and G67R showed partial loss. Surface electrostatics emphasised the significance of a positive patch on the binding surface of p16 that faces the CDK4 which was directly impacted due to mutations. Additionally, the partial binding mutants were found to have a lower stability compare to the Wildtype p16/CDK4 complex through the free energy landscape calculations. These findings provide useful insights into the molecular mechanisms by which p16 mutations influence CDK4 binding, potentially informing therapeutic strategies.
Collapse
Affiliation(s)
- D Arun
- Department of Biotechnology, National Institute of Technology Warangal, Hanamkonda, Telangana, India
| | - Soumya Lipsa Rath
- Department of Biotechnology, National Institute of Technology Warangal, Hanamkonda, Telangana, India.
| |
Collapse
|
10
|
Sarkar N, Kumar A. Paradigm shift: microRNAs interact with target gene promoters to cause transcriptional gene activation or silencing. Exp Cell Res 2025; 444:114372. [PMID: 39662662 DOI: 10.1016/j.yexcr.2024.114372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
MicroRNAs (miRNAs/miRs) are small (18-25 nucleotides in length), endogenous, non-coding RNAs that typically repress gene expression by interacting with the 3'untranslated regions (3'UTRs) of target mRNAs in the cytoplasm. While most of the scientific community still views miRNAs as repressors of gene expression, this review highlights their non-canonical novel role in the nucleus as activators or silencers of target gene transcription through miRNA-promoter interaction. The mechanistic details of the transcriptional role of miRNAs are yet to be elucidated, however, they can be explained by prospective models. In this review, we aim to discuss the different examples of transcriptional regulation by miRNAs and their possible mechanism of action, thereby offering a comprehensive perspective on the role of miRNAs in gene regulation and their importance in health and diseases.
Collapse
Affiliation(s)
- Neelanjana Sarkar
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| | - Arun Kumar
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
11
|
Laha S, Das S, Banerjee U, Ganguly T, Senapati S, Chatterjee G, Chatterjee R. Genome-wide RNA-seq, DNA methylation and small RNA-seq analysis unraveled complex gene regulatory networks in psoriasis pathogenesis. Gene 2025; 933:148903. [PMID: 39233195 DOI: 10.1016/j.gene.2024.148903] [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: 01/24/2024] [Revised: 08/12/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Psoriasis is a complex inflammatory skin disease characterized by reversible albeit relapsing red scaly plaques in the skin of a patient. In addition to the genetic predisposition, involvement of epigenetic and non-coding RNAs have also been liked with the disease. Nevertheless, any comprehensive study involving transcriptomic, small-RNA and DNA methylation at the genomic level from same patients is lacking. To investigate the complex regulation of molecular pathways in psoriasis, we carried out multi-omics integrative analysis of RNA-sequencing, small RNA-sequencing and DNA methylation profiling from the psoriatic and adjacent normal skin tissues. Our multi-omics analysis identified the genes and biological processes regulated either independently or in combination by DNA methylation and microRNAs. We identified miRNAs that specifically regulated keratinocyte hyper-proliferation, and cell cycle progression and checkpoint signaling in psoriasis. On contrary, DNA methylation was found to be more predominant in regulating immune and inflammatory responses, another causative factor in psoriasis pathogenesis. Many characteristic pathways in psoriasis e.g., Th17 cell differentiation and JAK-STAT signaling, were found to be regulated by both miRNAs and DNA methylation. We carried out functional characterization of a downregulated miRNA hsa-let-7c-5p, predicted to target upregulated genes in psoriasis involved in cell cycle processes, Th17 cell differentiation and JAK-STAT signaling pathways. Overexpression of hsa-let-7c-5p in keratinocytes caused the downregulation of its target genes, resulting in reduced cell proliferation and migration rates, demonstrating potential of miRNAs in regulating psoriasis pathogenesis. In conclusion, our findings identified distinct and shared gene-networks regulated by DNA methylation and miRNAs of a complex disease with reversible phenotype.
Collapse
Affiliation(s)
- Sayantan Laha
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Shantanab Das
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Urbee Banerjee
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Torsa Ganguly
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Swapan Senapati
- Consultant Dermatologist, Uttarpara, Hooghly, West Bengal 712258, India
| | - Gobinda Chatterjee
- Department of Dermatology, IPGMER/SSKM Hospital, Kolkata, West Bengal, India
| | - Raghunath Chatterjee
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India.
| |
Collapse
|
12
|
Mella C, Tsarouhas P, Brockwell M, Ball HC. The Role of Chronic Inflammation in Pediatric Cancer. Cancers (Basel) 2025; 17:154. [PMID: 39796780 PMCID: PMC11719864 DOI: 10.3390/cancers17010154] [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: 12/04/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Inflammation plays a crucial role in wound healing and the host immune response following pathogenic invasion. However, unresolved chronic inflammation can result in tissue fibrosis and genetic alterations that contribute to the pathogenesis of human diseases such as cancer. Recent scientific advancements exploring the underlying mechanisms of malignant cellular transformations and cancer progression have exposed significant disparities between pediatric and adult-onset cancers. For instance, pediatric cancers tend to have lower mutational burdens and arise in actively developing tissues, where cell-cycle dysregulation leads to gene, chromosomal, and fusion gene development not seen in adult-onset counterparts. As such, scientific findings in adult cancers cannot be directly applied to pediatric cancers, where unique mutations and inherent etiologies remain poorly understood. Here, we review the role of chronic inflammation in processes of genetic and chromosomal instability, the tumor microenvironment, and immune response that result in pediatric tumorigenesis transformation and explore current and developing therapeutic interventions to maintain and/or restore inflammatory homeostasis.
Collapse
Affiliation(s)
- Christine Mella
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
| | - Panogiotis Tsarouhas
- Department of Biology, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA;
| | - Maximillian Brockwell
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
| | - Hope C. Ball
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
| |
Collapse
|
13
|
Ntekoumes D, Song J, Liu H, Amelung C, Guan Y, Gerecht S. Acute Three-Dimensional Hypoxia Regulates Angiogenesis. Adv Healthc Mater 2025; 14:e2403860. [PMID: 39623803 PMCID: PMC11729260 DOI: 10.1002/adhm.202403860] [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: 10/06/2024] [Indexed: 01/15/2025]
Abstract
Hypoxia elicits a multitude of tissue responses depending on the severity and duration of the exposure. While chronic hypoxia is shown to impact development, regeneration, and cancer, the understanding of the threats of acute (i.e., short-term) hypoxia is limited mainly due to its transient nature. Here, a novel gelatin-dextran (Gel-Dex) hydrogel is established that decouples hydrogel formation and oxygen consumption and thus facilitates 3D sprouting from endothelial spheroids and, subsequently, induces hypoxia "on-demand." The Gel-Dex platform rapidly achieves acute moderate hypoxic conditions without compromising its mechanical properties. Acute exposure to hypoxia leads to increased endothelial cell migration and proliferation, promoting the total length and number of vascular sprouts. This work finds that the enhanced angiogenic response is mediated by reactive oxygen species, independently of hypoxia-inducible factors. Reactive oxygen species-dependent matrix metalloproteinases activity mediated angiogenic sprouting is observed following acute hypoxia. Overall, the Gel-Dex hydrogel offers a novel platform to study how "on-demand" acute moderate hypoxia impacts angiogenesis, with broad applicability to the development of novel sensing technologies.
Collapse
Affiliation(s)
- Dimitris Ntekoumes
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Haohao Liu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Connor Amelung
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Ya Guan
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
14
|
Nishida S, Konno T, Kohno T, Ohyanagi M, Nakano M, Ohwada K, Obata K, Kakuki T, Kakiuchi A, Kurose M, Takano K, Kojima T. Treatment with TNFα and lipolysis-stimulated lipoprotein receptor (LSR) antibody in the presence of HDAC inhibitors promotes apoptosis in human salivary duct adenocarcinoma. Tissue Barriers 2024:2437215. [PMID: 39676759 DOI: 10.1080/21688370.2024.2437215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024] Open
Abstract
Lipolysis-stimulated lipoprotein receptor (LSR), a lipid metabolism-related factor localized in tricellular tight junctions (tTJs), plays an important role in maintaining the epithelial homeostasis. LSR is highly expressed in well-differentiated cancers, and its expression decreases during malignancy. The LSR antibody inhibits cell growth and promotes apoptosis in some cancers. Histone deacetylases (HDACs) are thought to play a crucial role in carcinogenesis, and HDAC inhibitors promote differentiation and prevent cell proliferation and migration in cancers. HDAC inhibitors together with TNFα also induce apoptosis via TNFα-related apoptosis-inducing ligand (TRAIL) in some cancers. In this study, we investigated the apoptosis signaling induced by an anti-LSR antibody in human salivary duct adenocarcinoma (SDC) cell line A253, compared to TRAIL-induced apoptosis. A253 cells were treated with human recombinant TNFα with or without HDAC inhibitor trichostatin A (TSA) and quisinostat (JNJ-26481585). Treatment using TNFα with HDAC inhibitors markedly induced apoptosis in A253 cells and the anti-TNFα antibody prevented the induced apoptosis. A253 cells were treated with an antibody against the extracellular N-terminal domain of human LSR (LSR-N-ab) with or without HDAC inhibitors. Treatment with HDAC inhibitors induced LSR expression in the membranes of A253 cells. Treatment using LSR-N-ab with HDAC inhibitors markedly promoted apoptosis in A253 cells. The tricellular signaling pathway JNK inhibitor SP600125 and Hippo pathway MST1/2 inhibitor XMU-MP-1 prevented the apoptosis induced by treatment using TNFα or LSR-N-ab with HDAC inhibitors. Our findings indicated that treatment with TNFα or LSR-N-ab with HDAC inhibitors might be useful in the therapy for human SDC by enhancing apoptosis.
Collapse
Affiliation(s)
- Soshi Nishida
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takumi Konno
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kohno
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahiko Ohyanagi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaya Nakano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kizuku Ohwada
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazufumi Obata
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takuya Kakuki
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akito Kakiuchi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Kurose
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
15
|
Elmitwalli O, Darwish R, Al-Jabery L, Algahiny A, Roy S, Butler AE, Hasan AS. The Emerging Role of p21 in Diabetes and Related Metabolic Disorders. Int J Mol Sci 2024; 25:13209. [PMID: 39684919 DOI: 10.3390/ijms252313209] [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: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
In the context of cell cycle inhibition, anti-proliferation, and the dysregulation observed in certain cancer pathologies, the protein p21 assumes a pivotal role. p21 links DNA damage responses to cellular processes such as apoptosis, senescence, and cell cycle arrest, primarily functioning as a regulator of the cell cycle. However, accumulating empirical evidence suggests that p21 is both directly and indirectly linked to a number of different metabolic processes. Intriguingly, recent investigations indicate that p21 significantly contributes to the pathogenesis of diabetes. In this review, we present a comprehensive evaluation of the scientific literature regarding the involvement of p21 in metabolic processes, diabetes etiology, pancreatic function, glucose homeostasis, and insulin resistance. Furthermore, we provide an encapsulated overview of therapies that target p21 to alleviate metabolic disorders. A deeper understanding of the complex interrelationship between p21 and diabetes holds promise for informing current and future therapeutic strategies to address this rapidly escalating health crisis.
Collapse
Affiliation(s)
- Omar Elmitwalli
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Radwan Darwish
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Lana Al-Jabery
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ahmed Algahiny
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Sornali Roy
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Alexandra E Butler
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ammar S Hasan
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| |
Collapse
|
16
|
Magalhães M, Domínguez-Martín EM, Jorge J, Gonçalves AC, Massenzio F, Spigarelli R, Ribeiro-Rodrigues T, Catarino S, Girão H, Monti B, Spisni E, Ferreira L, Oliveira PJ, Efferth T, Rijo P, Cabral C. Unveiling the antitumor mechanism of 7α-acetoxy-6β-hydroxyroyleanone from Plectranthus hadiensis in glioblastoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118689. [PMID: 39128799 DOI: 10.1016/j.jep.2024.118689] [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: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Glioblastoma (GB) is the most aggressive and prevalent glioma within the central nervous system. Despite considerable efforts, GB continues to exhibit a dismal 5-year survival rate (∼6%). This is largely attributed to unfavorable prognosis and lack of viable treatment options. Therefore, novel therapies centered around plant-derived compounds emerge as a compelling avenue to enhance patient survival and well-being. The South African species, Plectranthus hadiensis Schweinf. (P. hadiensis), a member of the Lamiaceae family, has a history of use in traditional medicine for treating a range of diseases, including respiratory, digestive, and liver disorders. This species exhibits diverse biological activities, such as anti-inflammatory and antitumoral properties, likely attributed to its rich composition of naturally occurring diterpenes, like the abietane diterpene, 7α-acetoxy-6β-hydroxyroyleanone (Roy). Roy has demonstrated promising antitumor effects in various cancer cell lines, making it a compelling candidate for further investigation into its mechanisms against GB. AIM OF THE STUDY This study aims to investigate the antitumor activity and potential mechanism of Roy, a natural lead compound, in GB cells. MATERIAL AND METHODS Roy was isolated from the acetonic extract of P. hadiensis and its antitumor mechanism was assessed in a panel of human GB cell lines (U87, A172, H4, U373, and U118) to mimic tumor heterogeneity. Briefly, the impact of Roy treatment on the metabolic activity of cells was evaluated by Alamar Blue® assay, while cell death, cell cycle regulation, mitochondrial membrane potential, and activated caspase-3 activity were evaluated by flow cytometry. Measurement of mRNA levels of target genes was performed by qPCR, while protein expression was assessed by Western blotting. Cell uptake and impact on mitochondrial morphology were evaluated by confocal microscopy. RESULTS Roy induced G2/M cell cycle arrest, mitochondrial fragmentation, and apoptosis by inhibiting the expression of anti-apoptotic proteins and increasing the levels of activated caspase-3. The concentrations of Roy needed to achieve significant inhibitory outcomes were notably lower (6-9 fold) than those of temozolomide (TMZ), the standard first-line treatment, for achieving comparable effects. In addition, at low concentrations (16 μM), Roy affected the metabolic activity of tumor cells while having no significant impact on non-tumoral cells (microglia and astrocytes). CONCLUSION Overall, Roy demonstrated a robust antitumor activity against GB cells offering a promising avenue for the development of novel chemotherapeutic approaches.
Collapse
Affiliation(s)
- Mariana Magalhães
- University of Coimbra, Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Portugal; University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Eva María Domínguez-Martín
- CBIOS-Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal; Departamento de Ciencias Biomédicas, Facultad de Farmacia, Universidad de Alcalá de Henares, Madrid, Spain
| | - Joana Jorge
- University of Coimbra, Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, ICBR, Group of Environment Genetics and Oncobiology (CIMAGO)-Faculty of Medicine, Coimbra, Portugal
| | - Ana Cristina Gonçalves
- University of Coimbra, Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, ICBR, Group of Environment Genetics and Oncobiology (CIMAGO)-Faculty of Medicine, Coimbra, Portugal
| | - Francesca Massenzio
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Renato Spigarelli
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Teresa Ribeiro-Rodrigues
- University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Steve Catarino
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Henrique Girão
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Lino Ferreira
- University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; University of Coimbra, Faculty of Medicine, Coimbra, Portugal
| | - Paulo J Oliveira
- University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Patrícia Rijo
- CBIOS-Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal; Faculty of Pharmacy, Instituto de Investigação Do Medicamento (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Célia Cabral
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; University of Coimbra, Centre for Functional Ecology, Department of Life Sciences, Coimbra, Portugal.
| |
Collapse
|
17
|
Batista Brochado AC, de Moraes JA, Rodrigues de Oliveira B, De Souza Lima VH, Mariano ED, Karande S, Romasco T, Leite PEC, Mourão CF, Gomes Alves G. Metabolic and Regulatory Pathways Involved in the Anticancer Activity of Perillyl Alcohol: A Scoping Review of In Vitro Studies. Cancers (Basel) 2024; 16:4003. [PMID: 39682189 PMCID: PMC11640718 DOI: 10.3390/cancers16234003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Perillyl alcohol (POH), a plant-derived compound, has demonstrated anti-tumor activity across various human cancers. Understanding the regulatory pathways through which POH exerts its effects is crucial for identifying new therapeutic opportunities and exploring potential drug repositioning strategies. Therefore, this scoping review aims to provide a comprehensive overview of the metabolic and regulatory pathways involved in the anticancer effects of POH, based on in vitro evidence. METHODS Following the PRISMA-ScR 2018 guidelines, a systematic search was conducted in the PUBMED, Web of Science, and Scopus databases. RESULTS A total of 39 studies were included, revealing that POH exerts its biological effects by modulating several pathways, including the regulation of cyclins, CDKs, and p21, thereby affecting cell cycle progression. It inhibits growth and promotes cell death by attenuating AKT phosphorylation, reducing PARP-1 activity, increasing caspase activity and the FAS receptor and its ligand FASL. Additionally, POH reduces ERK phosphorylation, inhibits RAS protein isoprenylation, and decreases Na/K-ATPase activity. CONCLUSIONS In conclusion, this review delineates the key regulatory pathways responsible for mediating the biological effects of POH in cancer.
Collapse
Affiliation(s)
- Ana Carolina Batista Brochado
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | - Júlia Alves de Moraes
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| | - Bruna Rodrigues de Oliveira
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| | - Victor Hugo De Souza Lima
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | | | - Sachin Karande
- Dental Research Division, Department of Periodontology and Oral Implantology, Fluminense Federal University, Niteroi 21941-617, Brazil
| | - Tea Romasco
- Division of Dental Research Administration, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Paulo Emilio Correa Leite
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | - Carlos Fernando Mourão
- Department of Basic and Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Gutemberg Gomes Alves
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| |
Collapse
|
18
|
Malinowska K, Tarhonska K, Foksiński M, Sicińska P, Jabłońska E, Reszka E, Zarakowska E, Gackowski D, Górecka K, Balcerczyk A, Bukowska B. Impact of Short-Term Exposure to Non-Functionalized Polystyrene Nanoparticles on DNA Methylation and Gene Expression in Human Peripheral Blood Mononuclear Cells. Int J Mol Sci 2024; 25:12786. [PMID: 39684496 DOI: 10.3390/ijms252312786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The aim of the present study was to investigate the concentration- and size-dependent effects of non-functionalized polystyrene nanoparticles (PS-NPs) of varying diameters (29 nm, 44 nm, and 72 nm) on specific epigenetic modifications and gene expression profiles related to carcinogenesis in human peripheral blood mononuclear cells (PBMCs) in vitro. This in vitro human-cell-based model is used to investigate the epigenetic effect of various environmental xenobiotics. PBMCs were exposed to PS-NPs at concentrations ranging from 0.001 to 100 µg/mL for 24 h period. The analysis encompassed epigenetic DNA modifications, including levels of 5-methyl-2'-deoxycytidine (5-mdC) and 5-(hydroxymethyl)-2'-deoxycytidine (5-hmdC), as well as the levels of 2'-deoxyuridine (dU) and 5-(hydroxymethyl)-2'-deoxyuridine (5-hmdU) by mass spectrometry methods, methylation in the promoter regions of selected tumor suppressor genes TP53 (P53), CDKN2A (P16), and CDKN1A (P21) and proto-oncogenes (CCND1, BCL2, BCL6), along with the expression profile of the indicated genes by real-time PCR assays. The results obtained revealed no significant changes in global DNA methylation/demethylation levels in PBMCs after short-term exposure to non-functionalized PS-NPs. Furthermore, there were no changes observed in the level of dU, a product of cytosine deamination. However, the level of 5-hmdU, a product of both 5-hmdC deamination and thymine oxidation, was increased at the highest concentrations of larger PS-NPs (72 nm). None of the PS-NPs caused a change in the methylation pattern of the promoter regions of the TP53, CDKN2A, CDKN1A, CCND1, BCL2 and BCL6 genes. However, gene profiling indicated that PS-NPs with a diameter of 29 nm and 44 nm altered the expression of the TP53 gene. The smallest PS-NPs with a diameter of 29 nm increased the expression of the TP53 gene at a concentration of 10 µg/mL, while PS-NPs with a diameter of 44 nm did so at a concentration of 100 µg/mL. An increase in the expression of the CDKN2A gene was also observed when PBMCs were exposed to PS-NPs with 29 nm in diameter at the highest concentration. The observed effect depended on both the concentration and the size of the PS-NPs.
Collapse
Affiliation(s)
- Kinga Malinowska
- Department of Biophysics of Environmental Pollution, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland
| | - Kateryna Tarhonska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Teresy Str. 8, 91-348 Lodz, Poland
| | - Marek Foksiński
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland
| | - Paulina Sicińska
- Department of Biophysics of Environmental Pollution, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland
| | - Ewa Jabłońska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Teresy Str. 8, 91-348 Lodz, Poland
| | - Edyta Reszka
- Department of Biophysics of Environmental Pollution, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland
| | - Ewelina Zarakowska
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland
| | - Daniel Gackowski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland
| | - Karolina Górecka
- The Bio-Med-Chem Doctoral School, University of Lodz, 90-237 Lodz, Poland
- Lodz Institutes of the Polish Academy of Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Aneta Balcerczyk
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Bożena Bukowska
- Department of Biophysics of Environmental Pollution, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska Str. 141/143, 90-236 Lodz, Poland
| |
Collapse
|
19
|
Pathirana OC, Paranagama MP, Wijesundera KK, Mahakapuge TAN, Abeykoon AMAU, Rajapakse J. Elucidating the potential of Annona muricata L. grown in Sri Lanka to be used in developing an anticancer drug against colorectal and breast cancers. BMC Complement Med Ther 2024; 24:410. [PMID: 39609783 PMCID: PMC11603727 DOI: 10.1186/s12906-024-04712-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Since ancient times many traditional medicine systems around the world have been using different parts of Annona muricata L. (AM), to treat cancer. Indeed, numerous in vitro and in vivo studies also have shown anticancer properties of different solvent extracts of different parts of AM. Even the same part of the plant has shown different levels of anticancer properties based on geographical variations. Therefore, in the present study, the anticancer potential of the leaves, fruit pulp and the fruit peel of the AM that is grown in Sri Lanka was comparatively analyzed with the intention of identifying the most suitable part to be developed into a nutraceutical with anticancer effects. METHODS Freeze-dried aqueous extracts of immature leaves (ILAM), mature leaves (MLAM), pulp (PAM) and peel (PLAM) of AM were analyzed for their antioxidant activity using 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging and 2,2-azinobis-3-ethylbenzothiozoline-6-sulfonic acid (ABTS) cation decolorization assays. Their cytotoxicity on breast cancer (MCF-7) cells, colorectal adenocarcinoma (DLD-1) cells and normal human gingival fibroblasts (HGF-1) were determined by the 3-(4,5- dimethylthiazole-2-yl)-2,5-diphenyl-tetrazoliumbromide (MTT) assay. Their effect on mRNA expression of proapoptotic (Bax and caspase-7) and cell cycle arresting (p21) genes was analyzed by RT- qPCR in the same cell lines. RESULTS ILAM demonstrated the highest antioxidant activity in both DPPH and ABTS assays followed by MLAM, PLAM and PAM. In the MTT assay, both ILAM and MLAM demonstrated strong cytotoxic activity against MCF-7 and DLD-1 cell lines while there were no cytotoxic effects on the normal human cell line HGF-1. Both ILAM and MLAM demonstrated concentration-dependent upregulation of mRNA expression of cell cycle arresting gene p21 and apoptosis inducing genes Bax and caspase-7 in MCF-7 and DLD-1 cells. CONCLUSION The AEAM leaves grown in Sri Lanka has significantly higher antioxidant activity as well as selective cytotoxic effects on MCF-7 and DLD-1 cancer cells compared to its PL and P counterparts. Further, the AEAM leaves induced mRNA expression of the anticancer genes p21, Bax and caspase-7, indicating its potential to be developed into an anticancer drug against breast and colorectal cancer.
Collapse
Affiliation(s)
- Onela Canith Pathirana
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka.
| | - Madhavi Priyanka Paranagama
- Department of Basic Sciences, Faculty of Dental Sciences, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | - Kavindra Kumara Wijesundera
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | | | | | - Jayantha Rajapakse
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| |
Collapse
|
20
|
Almalki WH, Almujri SS. Aging, ROS, and cellular senescence: a trilogy in the progression of liver fibrosis. Biogerontology 2024; 26:10. [PMID: 39546058 DOI: 10.1007/s10522-024-10153-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
Ageing is an inevitable and multifaceted biological process that impacts a wide range of cellular and molecular mechanisms, leading to the development of various diseases, such as liver fibrosis. Liver fibrosis progresses to cirrhosis, which is an advanced form due to high amounts of extracellular matrix and restoration of normal liver structure with failure to repair damaged tissue and cells, marking the end of liver function and total liver failure, ultimately death. The most important factors are reactive oxygen species (ROS) and cellular senescence. Oxidative stress is defined as an impairment by ROS, which are by-products of the mitochondrial electron transport chain and other key molecular pathways that induce cell damage and can activate cellular senescence pathways. Cellular senescence is characterized by pro-inflammatory cytokines, growth factors, and proteases secreted by senescent cells, collectively known as the senescence-associated secretory phenotype (SASP). The presence of senescent cells, which disrupt tissue architecture and function and increase senescent cell production in liver tissues, contributes to fibrogenesis. Hepatic stellate cells (HSCs) are activated in response to chronic liver injury, oxidative stress, and senescence signals that drive excessive production and deposition of extracellular matrix. This review article aims to provide a comprehensive overview of the pathogenic role of ROS and cellular senescence in the aging liver and their contribution to fibrosis.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, 61421, Abha, Aseer, Saudi Arabia.
| |
Collapse
|
21
|
Basu A, Xuan Z. p21 Waf1/Cip1 Is a Novel Downstream Target of 40S Ribosomal S6 Kinase 2. Cancers (Basel) 2024; 16:3783. [PMID: 39594738 PMCID: PMC11592183 DOI: 10.3390/cancers16223783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: The ribosomal S6 kinase 2 (S6K2) acts downstream of the mechanistic target of rapamycin complex 1 and is a homolog of S6K1 but little is known about its downstream effectors. The objective of this study was to use an unbiased transcriptome profiling to uncover how S6K2 promotes breast cancer cell survival. Methods: RNA-Seq analysis was performed to identify novel S6K2 targets. Cells were transfected with siRNAs or plasmids containing genes of interest. Western blot analyses were performed to quantify total and phosphorylated proteins. Apoptosis was monitored by treating cells with different concentrations of doxorubicin. Results: Silencing of S6K2, but not S6K1, decreased p21 in MCF-7 and T47D breast cancer cells. Knockdown of Akt1 but not Akt2 decreased p21 in MCF-7 cells whereas both Akt1 and Akt2 knockdown attenuated p21 in T47D cells. While Akt1 overexpression enhanced p21 and partially reversed the effect of S6K2 deficiency on p21 downregulation in MCF-7 cells, it had little effect in T47D cells. S6K2 knockdown increased JUN mRNA and knockdown of cJun enhanced p21. Low concentrations of doxorubicin increased, and high concentrations decreased p21 levels in T47D cells. Silencing of S6K2 or p21 sensitized T47D cells to doxorubicin via c-Jun N-terminal kinase (JNK)-mediated downregulation of Mcl-1. Conclusions: S6K2 knockdown enhanced doxorubicin-induced apoptosis by downregulating the cell cycle inhibitor p21 and the anti-apoptotic protein Mcl-1 via Akt and/or JNK.
Collapse
Affiliation(s)
- Alakananda Basu
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Zhenyu Xuan
- Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, TX 75080, USA;
| |
Collapse
|
22
|
Zhang L, Lv Z, Xu QY, Wu B. TREM2 promotes the proliferation and invasion of renal cell carcinoma cells by inhibiting the P53 signaling pathway. Oncol Lett 2024; 28:538. [PMID: 39310025 PMCID: PMC11413725 DOI: 10.3892/ol.2024.14671] [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: 01/10/2024] [Accepted: 06/17/2024] [Indexed: 09/25/2024] Open
Abstract
Renal cell carcinoma (RCC) is a prevalent malignancy characterized by poor prognosis and high mortality. The role of triggering receptor expressed on myeloid cells-2 (TREM2) in RCC progression has been increasingly recognized, yet its underlying mechanisms remain to be fully elucidated. The aim of the present study was to assess the effects of TREM2 on RCC cells and its potential mechanisms. Lentiviral transfection was used to knockdown and overexpress TREM2 in RCC cells, and the expression level of TREM2 was evaluated using reverse transcription-quantitative PCR. Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assays were used to assess the proliferation of the RCC cells. Cell migration and invasion was evaluated using the wound healing assay and Transwell assay, respectively. Western blotting was used to assess the expression levels of TREM2, P53, p-P53, P21 and p-P21 in TREM2 knockdown or overexpression RCC cells. The results demonstrated that the expression level of TREM2 was significantly higher in cancer tissues compared with adjacent normal tissues. The results of the CCK-8 and EdU assays demonstrated that knockdown of TREM2 significantly inhibited the proliferation of RCC cells, whilst overexpression of TREM2 enhanced the proliferation of RCC cells. The results of the wound healing and Transwell assay revealed that, compared with the control group, the overexpression of TREM2 significantly increased the migration and invasion of RCC cells, whereas knockdown of TREM2 significantly decreased the migration of RCC cells. In addition, western blotting demonstrated that the phosphorylation levels of P53 and P21 proteins were significantly increased after TREM2 knockdown in RCC cells. In conclusion, TREM2 is highly expressed in RCC tissues and promotes the migration of RCC cells by inhibiting the P53 signaling pathway. The present study provides new insights into the regulatory effect of TREM2 on RCC and further reveals the potential of TREM2 as a therapeutic target for RCC.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Urology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213003, P.R. China
- Department of Urology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Zhong Lv
- Department of Urology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213003, P.R. China
- Department of Urology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Qin-Yu Xu
- Department of Urology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213003, P.R. China
- Department of Urology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Bin Wu
- Department of Urology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213003, P.R. China
- Department of Urology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
23
|
Thangavelu L, Altamimi ASA, Ghaboura N, Babu MA, Roopashree R, Sharma P, Pal P, Choudhary C, Prasad GVS, Sinha A, Balaraman AK, Rawat S. Targeting the p53-p21 axis in liver cancer: Linking cellular senescence to tumor suppression and progression. Pathol Res Pract 2024; 263:155652. [PMID: 39437639 DOI: 10.1016/j.prp.2024.155652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Liver cancer is a major health epidemic worldwide, mainly due to its high mortality rates and limited treatment options. The association of cellular senescence to tumorigenesis and the cancer hallmarks remains a subject of interest in cancer biology. The p53-p21 signalling axis is an important regulator in restoring the cell's balance by supporting tumor suppression and tumorigenesis in liver cancer. We review the novel molecular mechanisms that p53 and its downstream effector, p21, employ to induce cellular senescence, making it last longer, and halt the proliferation of damaged hepatocytes to become tumorous cells. We also examine how dysregulation of this pathway contributes to HCC pathogenesis, proliferation, survival, acquired resistance to apoptosis, and increased invasiveness. Furthermore, we comprehensively describe the molecular cross-talk between the p53-p21 signalling axis and major cell cycle signalling pathways, including Wnt/β-catenin, PI3K/Akt, and TGF-β in liver cancer and provide an overview of promising candidates for chemoprevention and future therapeutic strategies. This review article explores the roles of the p53-p21 pathway in liver cancer, examining its function in promoting cellular senescence under normal conditions and its potential role in cancer progression. It also highlights novel therapeutic drugs and drug targets within the pathway and discusses the implications for treatment strategies and prognosis in liver cancer.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Pusparghya Pal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Chhavi Choudhary
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| |
Collapse
|
24
|
Khan AQ, Al-Tamimi M, Anver R, Agha MV, Anamangadan G, Raza SS, Ahmad F, Ahmad A, Alam M, Buddenkotte J, Steinhoff M, Uddin S. Targeting of S-phase kinase associated protein 2 stabilized tumor suppressors leading to apoptotic cell death in squamous skin cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167286. [PMID: 38866114 DOI: 10.1016/j.bbadis.2024.167286] [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: 11/21/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
S-phase kinase-associated protein 2 (Skp2) is an F-box protein overexpressed in human cancers and linked with poor prognosis. It triggers cancer pathogenesis, including stemness and drug resistance. In this study, we have explored the potential role of Skp2 targeting in restoring the expression of tumor suppressors in human cutaneous squamous cell carcinoma (cSCC) cells. Our results showed that genetic and pharmacological Skp2 targeting markedly suppressed cSCC cell proliferation, colony growth, spheroid formation, and enhanced sensitization to chemotherapeutic drugs. Further, western blot results demonstrated restoration of tumor suppressor (KLF4) and CDKI (p21) and suppression of vimentin and survivin in Skp2-knocked-down cSCC cells. Importantly, we also explored that Skp2 targeting potentiates apoptosis of cSCC cells through MAPK signaling. Moreover, co-targeting of Skp2 and PI3K/AKT resulted in increased cancer cell death. Interestingly, curcumin, a well-known naturally derived anticancer agent, also inhibits Skp2 expression with concomitant CDKI upregulation. In line, curcumin suppressed cSCC cell growth through ROS-mediated apoptosis, while the use of N-acetyl cysteine (NAC) reversed curcumin-induced cell death. Curcumin treatment also sensitized cSCC cells to conventional anticancer drugs, such as cisplatin and doxorubicin. Altogether, these data suggest that Skp2 targeting restores the functioning of tumor suppressors, inhibits the expression of genes associated with cell proliferation and stemness, and sensitizes cancer cells to anticancer drugs. Thus, genetic, and pharmacological ablation of Skp2 can be an important strategy for attenuating cancer pathogenesis and associated complications in skin squamous cell carcinoma.
Collapse
Affiliation(s)
- Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Maha Al-Tamimi
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Rasheeda Anver
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Maha Victor Agha
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Gazala Anamangadan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow 226003, India
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar; Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; College of Medicine, Qatar University, Doha 2713, Qatar.
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar.
| |
Collapse
|
25
|
Jia X, Zhao Y, Li Q, Lu X, Wang X, Wang H, Shi Z, Xu Y, Huang B, Huang F, Wang Y. Targeted Inhibition of p21 Promotes the Growth of Breast Cancer Cells and Impairs the Tumor-Killing Effect of the Vaccinia Virus. J Breast Cancer 2024; 27:293-304. [PMID: 39344409 PMCID: PMC11543277 DOI: 10.4048/jbc.2024.0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/18/2024] [Accepted: 08/15/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE Vaccinia virus is widely used as an oncolytic agent for human cancer therapy, and several versions of vaccinia virus have demonstrated robust antitumor effects in breast cancer. Most vaccinia viruses are modified by thymidine kinase (TK) deletion. The function of the cyclin-dependent kinase inhibitor p21 in breast cancer remains controversial. We explored the impact of p21 gene knockdown (KD) on breast cancer cells and whether p21 KD interferes with the antitumor effect of TK-negative vaccinia virus. METHODS p21 KD MDA-MB-231 and p21 KD MCF-7 cells were prepared, and cell proliferation and migration rates were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and scratch healing assays. The tumor growth of xenografts originating from p21KD MDA-MB-231 cells and control cells was compared in a mouse model. The colony formation and sphere-forming abilities of p21 KD breast cancer cells were also determined using low-melting agarose and serum-free culture. The tumor-killing effect of the vaccinia virus was determined in breast cancer cells and mouse models using an MTT assay and tumor cell xenografts. RESULTS p21 KD increased the growth and migration of MDA-MB-231 and MCF-7 cells and promoted the cell growth of MDA-MB-231 cells in mice, while decreasing the colony formation and sphere formation abilities. Expression of TK was reduced in p21 KD MDA-MB-231 cells. Oncolytic effects of both wild-type and TK-deleted vaccinia viruses were attenuated in p21KD MDA-MB-231 cells. The tumor-killing effect of TK-deleted vaccinia virus was also weakened in xenografted mice bearing p21 KD MDA-MB-231 cells. CONCLUSION Targeted inhibition of p21 accelerates the proliferation and migration of breast cancer cells and impairs the tumor-killing effect of vaccinia virus, suggesting that p21 levels in cancer cells interfere with vaccinia virus oncolytic therapy.
Collapse
Affiliation(s)
- Xiaoyuan Jia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yujia Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qiang Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaming Lu
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Xiaoyan Wang
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
| | - Hui Wang
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
| | - Ziyi Shi
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yipeng Xu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fang Huang
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| |
Collapse
|
26
|
Bhattacharya R, Kumari J, Banerjee S, Tripathi J, Parihar SS, Mohan N, Sinha P. Hippo effector, Yorkie, is a tumor suppressor in select Drosophila squamous epithelia. Proc Natl Acad Sci U S A 2024; 121:e2319666121. [PMID: 39288176 PMCID: PMC11441523 DOI: 10.1073/pnas.2319666121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Mammalian Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) and Drosophila Yorkie (Yki) are transcription cofactors of the highly conserved Hippo signaling pathway. It has been long assumed that the YAP/TAZ/Yki signaling drives cell proliferation during organ growth. However, its instructive role in regulating developmentally programmed organ growth, if any, remains elusive. Out-of-context gain of YAP/TAZ/Yki signaling often turns oncogenic. Paradoxically, mechanically strained, and differentiated squamous epithelia display developmentally programmed constitutive nuclear YAP/TAZ/Yki signaling. The unknown, therefore, is how a growth-promoting YAP/TAZ/Yki signaling restricts proliferation in differentiated squamous epithelia. Here, we show that reminiscent of a tumor suppressor, Yki negatively regulates the cell growth-promoting PI3K/Akt/TOR signaling in the squamous epithelia of Drosophila tubular organs. Thus, downregulation of Yki signaling in the squamous epithelium of the adult male accessory gland (MAG) up-regulates PI3K/Akt/TOR signaling, inducing cell hypertrophy, exit from their cell cycle arrest, and, finally, culminating in squamous cell carcinoma (SCC). Thus, blocking PI3K/Akt/TOR signaling arrests Yki loss-induced MAG-SCC. Further, MAG-SCCs, like other lethal carcinomas, secrete a cachectin, Impl2-the Drosophila homolog of mammalian IGFBP7-inducing cachexia and shortening the lifespan of adult males. Moreover, in the squamous epithelium of other tubular organs, like the dorsal trunk of larval tracheal airways or adult Malpighian tubules, downregulation of Yki signaling triggers PI3K/Akt/TOR-induced cell hypertrophy. Our results reveal that Yki signaling plays an instructive, antiproliferative role in the squamous epithelia of tubular organs.
Collapse
Affiliation(s)
- Rachita Bhattacharya
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Jaya Kumari
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Shweta Banerjee
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Jyoti Tripathi
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Saurabh Singh Parihar
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Nitin Mohan
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Pradip Sinha
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| |
Collapse
|
27
|
Myeza N, Slabber C, Munro OQ, Sookai S, Zacharias SC, Martins-Furness C, Harmse L. An 8-aminoquinoline-naphthyl copper complex causes apoptotic cell death by modulating the expression of apoptotic regulatory proteins in breast cancer cells. Eur J Pharmacol 2024; 978:176764. [PMID: 38908670 DOI: 10.1016/j.ejphar.2024.176764] [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: 02/27/2024] [Revised: 06/08/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Breast cancer is one of the most common cancers globally and a leading cause of cancer-related deaths among women. Despite the combination of chemotherapy with targeted therapy, including monoclonal antibodies and kinase inhibitors, drug resistance and treatment failure remain a common occurrence. Copper, complexed to various organic ligands, has gained attention as potential chemotherapeutic agents due to its perceived decreased toxicity to normal cells. The cytotoxic efficacy and the mechanism of cell death of an 8-aminoquinoline-naphthyl copper complex (Cu8AqN) in MCF-7 and MDA-MB-231 breast cancer cell lines was investigated. The complex inhibited the growth of MCF-7 and MDA-MB-231 cells with IC50 values of 2.54 ± 0.69 μM and 3.31 ± 0.06 μM, respectively. Nuclear fragmentation, annexin V binding, and increased caspase-3/7 activity indicated apoptotic cell death. The loss of mitochondrial membrane potential, an increase in caspase-9 activity, the absence of active caspase-8 and a decrease of tumour necrosis factor receptor 1(TNFR1) expression supported activation of the intrinsic apoptotic pathway. Increased ROS formation and increased expression of haem oxygenase-1 (HMOX-1) indicated activation of cellular stress pathways. Expression of p21 protein in the nuclei was increased indicating cell cycle arrest, whilst the expression of inhibitor of apoptosis proteins (IAPs); cIAP1, XIAP and survivin were decreased, creating a pro-apoptotic environment. Phosphorylated p53 species; phospho-p53(S15), phospho-p53(S46), and phospho-p53(S392) accumulated in MCF-7 cells indicating the potential of Cu8AqN to restore p53 function in the cells. In combination, the data indicates that Cu8AqN is a useful lead molecule worthy of further exploration as a potential anti-cancer drug.
Collapse
Affiliation(s)
- Nonzuzo Myeza
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa
| | - Cathy Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| | - Sheldon Sookai
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg, 2017, South Africa
| | - Savannah C Zacharias
- School of Chemistry and Physics, University of KwaZulu-Natal, King Edward Drive, Pietermaritzburg, Scottsville, 3209, South Africa
| | - Carla Martins-Furness
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa
| | - Leonie Harmse
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, South Africa.
| |
Collapse
|
28
|
Ran X, Hu A, Kuang Y, Wang C, Liu W, Xiao X, Zacksenhaus E, Yu X, Ben-David Y. UM171 suppresses breast cancer progression by inducing KLF2. Breast Cancer Res Treat 2024; 207:405-415. [PMID: 38874684 PMCID: PMC11297059 DOI: 10.1007/s10549-024-07372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/09/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE Breast cancer is the most frequent cancer in women with significant death rate. Morbidity is associated with drug resistance and metastasis. Development of novel drugs is unmet need. The aim of this study is to show potent anti-neoplastic activity of the UM171 compound on breast cancer cells and its mechanism of action. METHODS The inhibitory effect of UM171 on several breast cancer (BC) cell lines was examined using MTT and colony-forming assays. Cell cycle and apoptosis assays were utilized to determine the effect of UM171 on BC cell proliferation and survival. Wound healing scratch and transwell migration assays were used to examine the migration of BC cell lines in culture. Xenograft of mouse model with 4T1 cells was used to determine inhibitory effect of UM171 in vivo. Q-RT-PCR and western blotting were used to determine the expression level of genes effected by UM171. Lentivirus-mediated shRNAs were used to knockdown the expression of KLF2 in BC cells. RESULTS UM171 was previously identified as a potent agonist of human hematopoietic stem cell renewal and inhibitor of leukemia. In this study, UM171 was shown to inhibit the growth of multiple breast cancer cell lines in culture. UM171-mediated growth inhibition was associated with the induction of apoptosis, G2/M cell cycle arrest, lower colony-forming capacity, and reduced motility. In a xenotransplantation model of mouse triple-negative breast cancer 4T1 cells injected into syngeneic BALB/c mice, UM171 strongly inhibited tumor growth at a level comparable to control paclitaxel. UM171 increased the expression of the three PIM genes (PIM1-3) in breast cancer cells. Moreover, UM171 strongly induced the expression of the tumor suppressor gene KLF2 and cell cycle inhibitor P21CIP1. Accordingly, knockdown of KLF2 using lentivirus-mediated shRNA significantly attenuated the growth suppressor activity of UM171. As PIM1-3 act as oncogenes and are involved in breast cancer progression, induction of these kinases likely impedes the inhibitory effect of KLF2 induction by UM171. Accordingly, combination of UM171 with a PAN-PIM inhibitor LGH447 significantly reduced tumor growth in culture. CONCLUSION These results suggested that UM171 inhibited breast cancer progression in part through activation of KLF2 and P21. Combination of UM171 with a PAN-PIM inhibitor offer a novel therapy for aggressive forms of breast cancer.
Collapse
Affiliation(s)
- Xiaojuan Ran
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou, People's Republic of China
- The Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, People's Republic of China
| | - Yi Kuang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou, People's Republic of China
- The Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, People's Republic of China
| | - Chunlin Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou, People's Republic of China
- The Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, People's Republic of China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou, People's Republic of China
- The Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, People's Republic of China
| | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou, People's Republic of China
- The Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, People's Republic of China
| | - Eldad Zacksenhaus
- Department of Medicine, Laboratory Medicine & Pathobiology and Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L1, Canada
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Xiangdi Yu
- Guizhou University of Traditional Chinese Medicine, Guiyang, China.
- Anesthesiology Department of Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, 545000, Guangxi, China.
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou, People's Republic of China.
- The Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, People's Republic of China.
| |
Collapse
|
29
|
Gu W, Wang M, Wu Z, Cai Y. Ubiquitination of p21 by E3 ligase RNF135 promotes the proliferation of human glioblastoma cells. Oncol Lett 2024; 28:406. [PMID: 38988442 PMCID: PMC11234804 DOI: 10.3892/ol.2024.14539] [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: 03/20/2024] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a highly heterogeneous tumor of the central nervous system with a high mortality rate. The upregulation of RING finger protein 135 (RNF135), an E3 ligase, has been observed in GBM, but the associated mechanisms have not been fully elucidated. The aim of the present study was to identify the substrate of RNF135 and study its functions in GBM. Bioinformatics analyses were performed. In addition, RNF135 was overexpressed or knocked down in human U87 and U251 GBM cells, and the effect on cell proliferation was analyzed using Cell Counting Kit-8 and colony formation assays. Furthermore, the interaction of RNF135 with its potential substrate was analyzed using glutathione S-transferase, yeast two-hybrid, immunoprecipitation (IP), co-IP and immunoblotting assays. Bioinformatics analysis indicated that RNF135 serves as a marker of poor prognosis in GBM. The overexpression of RNF135 was demonstrated to promote the proliferation of GBM cells, while the knockdown of RNF135 inhibited GBM cell growth. In addition, the results of the interaction experiments indicate that RNF135 interacts with p21 and mediates the degradation of p21 by ubiquitination. The major site of RNF135-mediated p21 ubiquitination was identified as K163. Further experiments demonstrated that RNF135 promotes the proliferation of GBM cells mainly via p21. In summary, these findings suggest that RNF135 has potential as a therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Weiting Gu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Ming Wang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Zhebao Wu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Yu Cai
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
30
|
Kang M, Lee Y, Lee Y, Kim E, Jo J, Shin H, Choi J, Oh J, Yoon H, Kang HW. Wavelength-dependent photobiomodulation (PBM) for proliferation and angiogenesis of melanoma tumor in vitro and in vivo. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:112990. [PMID: 39032372 DOI: 10.1016/j.jphotobiol.2024.112990] [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: 05/17/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Photobiomodulation (PBM) has widely been used to effectively treat complications associated with cancer treatment, including oral mucositis, radiation dermatitis, and surgical wounds. However, the safety of PBM against cancer still needs to be validated as the effects of PBM on cancer cells and their mechanisms are unclear. The current study investigated the wavelength-dependent PBM effects by examining four different laser wavelengths (405, 532, 635, and 808 nm) on B16F10 melanoma tumor cells. In vitro tests showed that PBM with 808 nm promoted both proliferation and migration of B16F10 cells. In vivo results demonstrated that PBM with 808 nm significantly increased the relative tumor volume and promoted angiogenesis with overexpression of VEGF and HIF-1α. In addition, PBM induced the phosphorylation of factors closely related to cancer cell proliferation and tumor growth and upregulated the related gene expression. The current result showed that compared to the other wavelengths, 808 nm yielded a significant tumor-stimulating effect the malignant melanoma cancer. Further studies will investigate the in-depth molecular mechanism of PBM on tumor stimulation in order to warrant the safety of PBM for clinical cancer treatment.
Collapse
Affiliation(s)
- Myungji Kang
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
| | - Yeachan Lee
- Center for Advanced Models for Translational Sciences and Therapeutics and Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuri Lee
- School of Life Science, Handong Global University, Pohang 37554, Republic of Korea
| | - Eunjung Kim
- School of Life Science, Handong Global University, Pohang 37554, Republic of Korea
| | - Jihye Jo
- Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare and Digital Healthcare Research Center, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea
| | - Hwarang Shin
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
| | - Jaeyeop Choi
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Junghwan Oh
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare and Digital Healthcare Research Center, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea; Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Hongsup Yoon
- School of Life Science, Handong Global University, Pohang 37554, Republic of Korea.
| | - Hyun Wook Kang
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare and Digital Healthcare Research Center, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
31
|
Chang S, Ren D, Zhang L, Liu S, Yang W, Cheng H, Zhang X, Hong E, Geng D, Wang Y, Chen C, Zhang J, Shi T, Guo Y, Ni X, Wang H, Jin Y. Therapeutic SHPRH-146aa encoded by circ-SHPRH dynamically upregulates P21 to inhibit CDKs in neuroblastoma. Cancer Lett 2024; 598:217120. [PMID: 39002691 DOI: 10.1016/j.canlet.2024.217120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Recent research has underscored the significance of circular RNAs (circRNAs) in various cancers, including neuroblastoma (NB). Specifically, circ-SHPRH, a unique circRNA, has been revealed to inhibit tumor growth by sequestering miRNAs or producing the SHPRH-146aa protein. To explore circ-SHPRH's involvement in NB and its potential application in gene therapy, this study examined circ-SHPRH expression in 94 NB tissues and cell lines (SK-N-BE(2), SH-SY5Y) using real-time PCR and fluorescence in situ hybridization (FISH). Functional assays encompassing both overexpression and knockdown experiments in NB cell lines, as well as in vivo investigations, were conducted. RNA-seq analysis revealed a correlation between circ-SHPRH and the pathway of P21 (CDKN1A), a pivotal cell cycle regulator. Validation through PCR and other techniques confirmed that circ-SHPRH upregulated P21 expression. Furthermore, the regulatory role of circ-SHPRH in the P21-CDK pathway was corroborated through SHPRH-146aa expression analysis. Notably, adenovirus-mediated circ-SHPRH overexpression effectively curbed NB tumor growth in NSG mice, while combining circ-SHPRH with everolimus exhibited potential for NB treatment. This study elucidates the remarkable significance of circ-SHPRH in NB and its prospective utility in gene therapy, thereby paving the way for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Saishuo Chang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Dong Ren
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Li Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shan Liu
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Wei Yang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Haiyan Cheng
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xuexi Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Enyu Hong
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Di Geng
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children's Hospital, National Center for Children's Health, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yadi Wang
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children's Hospital, National Center for Children's Health, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Chenghao Chen
- Department of Thoracic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Tieliu Shi
- Center for Bioinformatics and Computational Biology and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yongli Guo
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China; Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Huanmin Wang
- Department of Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
32
|
Wei Y, Mou S, Yang Q, Liu F, Cooper ME, Chai Z. To target cellular senescence in diabetic kidney disease: the known and the unknown. Clin Sci (Lond) 2024; 138:991-1007. [PMID: 39139135 PMCID: PMC11327223 DOI: 10.1042/cs20240717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/07/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024]
Abstract
Cellular senescence represents a condition of irreversible cell cycle arrest, characterized by heightened senescence-associated beta-galactosidase (SA-β-Gal) activity, senescence-associated secretory phenotype (SASP), and activation of the DNA damage response (DDR). Diabetic kidney disease (DKD) is a significant contributor to end-stage renal disease (ESRD) globally, with ongoing unmet needs in terms of current treatments. The role of senescence in the pathogenesis of DKD has attracted substantial attention with evidence of premature senescence in this condition. The process of cellular senescence in DKD appears to be associated with mitochondrial redox pathways, autophagy, and endoplasmic reticulum (ER) stress. Increasing accumulation of senescent cells in the diabetic kidney not only leads to an impaired capacity for repair of renal injury, but also the secretion of pro-inflammatory and profibrotic cytokines and growth factors causing inflammation and fibrosis. Current treatments for diabetes exhibit varying degrees of renoprotection, potentially via mitigation of senescence in the diabetic kidney. Targeting senescent cell clearance through pharmaceutical interventions could emerge as a promising strategy for preventing and treating DKD. In this paper, we review the current understanding of senescence in DKD and summarize the possible therapeutic interventions relevant to senescence in this field.
Collapse
Affiliation(s)
- Yuehan Wei
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, Australia
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Mou
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Ren Ji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Yang
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Liu
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Mark E Cooper
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Zhonglin Chai
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
33
|
Algranati D, Oren R, Dassa B, Fellus-Alyagor L, Plotnikov A, Barr H, Harmelin A, London N, Ron G, Furth N, Shema E. Dual targeting of histone deacetylases and MYC as potential treatment strategy for H3-K27M pediatric gliomas. eLife 2024; 13:RP96257. [PMID: 39093942 PMCID: PMC11296706 DOI: 10.7554/elife.96257] [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] [Indexed: 08/04/2024] Open
Abstract
Diffuse midline gliomas (DMGs) are aggressive and fatal pediatric tumors of the central nervous system that are highly resistant to treatments. Lysine to methionine substitution of residue 27 on histone H3 (H3-K27M) is a driver mutation in DMGs, reshaping the epigenetic landscape of these cells to promote tumorigenesis. H3-K27M gliomas are characterized by deregulation of histone acetylation and methylation pathways, as well as the oncogenic MYC pathway. In search of effective treatment, we examined the therapeutic potential of dual targeting of histone deacetylases (HDACs) and MYC in these tumors. Treatment of H3-K27M patient-derived cells with Sulfopin, an inhibitor shown to block MYC-driven tumors in vivo, in combination with the HDAC inhibitor Vorinostat, resulted in substantial decrease in cell viability. Moreover, transcriptome and epigenome profiling revealed synergistic effect of this drug combination in downregulation of prominent oncogenic pathways such as mTOR. Finally, in vivo studies of patient-derived orthotopic xenograft models showed significant tumor growth reduction in mice treated with the drug combination. These results highlight the combined treatment with PIN1 and HDAC inhibitors as a promising therapeutic approach for these aggressive tumors.
Collapse
Affiliation(s)
- Danielle Algranati
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Roni Oren
- Department of Veterinary Resources, Weizmann Institute of ScienceRehovotIsrael
| | - Bareket Dassa
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Faculty of Biochemistry, Weizmann Institute of ScienceRehovotIsrael
| | - Liat Fellus-Alyagor
- Department of Veterinary Resources, Weizmann Institute of ScienceRehovotIsrael
| | - Alexander Plotnikov
- Wohl Institute for Drug Discovery of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of ScienceRehovotIsrael
| | - Haim Barr
- Wohl Institute for Drug Discovery of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of ScienceRehovotIsrael
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of ScienceRehovotIsrael
| | - Nir London
- Department of Chemical and Structural Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Guy Ron
- Racah Institute of Physics, Hebrew UniversityJerusalemIsrael
| | - Noa Furth
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Efrat Shema
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
34
|
Gu T, Wen Y, Zhou Q, Yuan W, Guo H, Chang WL, Yang Q. Fungal metabolite altersolanol a exhibits potent cytotoxicity against human placental trophoblasts in vitro via mitochondria-mediated apoptosis. Mycotoxin Res 2024; 40:419-432. [PMID: 38717551 DOI: 10.1007/s12550-024-00539-0] [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: 01/08/2024] [Revised: 03/24/2024] [Accepted: 04/29/2024] [Indexed: 07/19/2024]
Abstract
Altersolanol A, a fungus-derived tetrahydroanthraquinone, has shown cytotoxic effects on multiple cancer cells. However, its reproductive toxicity in humans has not been well-addressed. The present study was aimed at investigating the cytotoxicity of altersolanol A on human placental trophoblasts including choriocarcinoma cell line JEG-3 and normal trophoblast cell line HTR-8/SVneo in vitro. The results showed that altersolanol A inhibited proliferation and colony formation of human trophoblasts, and the choriocarcinoma cells were more sensitive to the compound than the normal trophoblasts. Altersolanol A induced cell cycle arrest at G2/M phase in JEG-3 cells and S phase in HTR-8/SVneo cells, downregulated the expression of cell cycle-related checkpoint proteins, and upregulated the p21 level. Altersolanol A also promoted apoptosis in human trophoblasts via elevating the Bax/Bcl-2 ratio and decreasing both caspase-3 and caspase-9 levels. Meanwhile, altersolanol A suppressed the mitochondrial membrane potential and induced ROS production and cytochrome c release, which activated the mitochondria-mediated intrinsic apoptosis. Moreover, migration and invasion were inhibited upon altersolanol A exposure with downregulation of matrix metalloproteinase (MMP)-2 in JEG-3 cells and MMP-9 in HTR-8/SVneo cells. Mechanically, altersolanol A supplement decreased the phosphorylation of JNK, ERK, and p38, manifesting the inactivation of MAPK signaling pathway in the human trophoblasts. In conclusion, altersolanol A exhibited potential reproductive cytotoxicity against human trophoblasts via promoting mitochondrial-mediated apoptosis and inhibiting the MAPK signaling pathway.
Collapse
Affiliation(s)
- Ting Gu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Yuting Wen
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Qian Zhou
- Hunan Provincial Key Laboratory for Biology and , Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha, 410128, China
| | - Wei Yuan
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Department of Obstetrics, Affiliated Longhua People's Hospital, Southern Medical University (Longhua People's Hospital), Shenzhen, 518036, China
| | - Haichun Guo
- Changsha Hospital for Maternal & Child Health Care of Hunan Normal University, Changsha, 410007, China
| | - Wen-Lin Chang
- Department of Obstetrics, Affiliated Longhua People's Hospital, Southern Medical University (Longhua People's Hospital), Shenzhen, 518036, China.
| | - Qing Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
35
|
Lu Y, Li L, Li J, Wang M, Yang J, Zhang M, Jiang Q, Tang X. Prx1/PHB2 axis mediates mitophagy in oral leukoplakia cellular senescence. Pathol Res Pract 2024; 260:155411. [PMID: 38936092 DOI: 10.1016/j.prp.2024.155411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Oral leukoplakia (OLK) is the most common oral potentially malignant disorder (OPMD), which can be malignantly transformed into oral squamous cell carcinoma (OSCC). Peroxiredoxin1(Prx1) has been predicted to bind to Prohibitin2 (PHB2), which confers to affect OLK progression; however, the mechanism of Prx1/PHB2 mediated mitophagy involved in OLK remains unclear. METHODS This study aimed to explore the mechanism of the Prx1/PHB2 axis on senescence in OLK through mediating mitophagy. The positive rate of Ki67 and the expression of p21, p16, PHB2, and LC3 in human normal, OLK, and OSCC tissues were detected by immunohistochemical staining. The mitophagy and mitochondrial function changes were then analyzed in Prx1 knockdown and Prx1C52S mutations in dysplastic oral keratinocyte (DOK) cells treated with H2O2. In situ Proximity Ligation Assay combined with co-immunoprecipitation was used to detect the interaction between Prx1 and PHB2. RESULTS Clinically, the positive rate of Ki67 progressively increased from normal to OLK, OLK with dysplasia, and OSCC. Higher p21, p16, PHB2, and LC3 expression levels were observed in OLK with dysplasia than in normal and OSCC tissues. In vitro, PHB2 and LC3II expression gradually increased with the degree of DOK cell senescence. Prx1/PHB2 regulated mitophagy and affected senescence in H2O2-induced DOK cells. Furthermore, Prx1C52S mutation specifically reduced interaction between Prx1 and PHB2. Prx1Cys52 is associated with mitochondrial reactive oxygen species (ROS) accumulated and cell cycle arrest. CONCLUSION Prx1Cys52 functions as a redox sensor that binds to PHB2 and regulates mitophagy in the senescence of OLK, suggesting its potential as a clinical target.
Collapse
Affiliation(s)
- Yunping Lu
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Lingyu Li
- Division of Oral Pathology, Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Jing Li
- Division of Oral Pathology, Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Min Wang
- Division of Oral Pathology, Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Jing Yang
- Division of Oral Pathology, Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Min Zhang
- Division of Oral Pathology, Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Qingsong Jiang
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Xiaofei Tang
- Division of Oral Pathology, Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
36
|
Kafida M, Karela M, Giakountis A. RNA-Independent Regulatory Functions of lncRNA in Complex Disease. Cancers (Basel) 2024; 16:2728. [PMID: 39123456 PMCID: PMC11311644 DOI: 10.3390/cancers16152728] [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: 07/06/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
During the metagenomics era, high-throughput sequencing efforts both in mice and humans indicate that non-coding RNAs (ncRNAs) constitute a significant fraction of the transcribed genome. During the past decades, the regulatory role of these non-coding transcripts along with their interactions with other molecules have been extensively characterized. However, the study of long non-coding RNAs (lncRNAs), an ncRNA regulatory class with transcript lengths that exceed 200 nucleotides, revealed that certain non-coding transcripts are transcriptional "by-products", while their loci exert their downstream regulatory functions through RNA-independent mechanisms. Such mechanisms include, but are not limited to, chromatin interactions and complex promoter-enhancer competition schemes that involve the underlying ncRNA locus with or without its nascent transcription, mediating significant or even exclusive roles in the regulation of downstream target genes in mammals. Interestingly, such RNA-independent mechanisms often drive pathological manifestations, including oncogenesis. In this review, we summarize selective examples of lncRNAs that regulate target genes independently of their produced transcripts.
Collapse
Affiliation(s)
| | | | - Antonis Giakountis
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, Mezourlo, 41500 Larissa, Greece
| |
Collapse
|
37
|
Visuddho V, Halim P, Helen H, Muhar AM, Iqhrammullah M, Mayulu N, Surya R, Tjandrawinata RR, Ribeiro RIMA, Tallei TE, Taslim NA, Kim B, Syahputra RA, Nurkolis F. Modulation of Apoptotic, Cell Cycle, DNA Repair, and Senescence Pathways by Marine Algae Peptides in Cancer Therapy. Mar Drugs 2024; 22:338. [PMID: 39195454 DOI: 10.3390/md22080338] [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: 05/25/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Marine algae, encompassing both macroalgae and microalgae, have emerged as a promising and prolific source of bioactive compounds with potent anticancer properties. Despite their significant therapeutic potential, the clinical application of these peptides is hindered by challenges such as poor bioavailability and susceptibility to enzymatic degradation. To overcome these limitations, innovative delivery systems, particularly nanocarriers, have been explored. Nanocarriers, including liposomes, nanoparticles, and micelles, have demonstrated remarkable efficacy in enhancing the stability, solubility, and bioavailability of marine algal peptides, ensuring controlled release and prolonged therapeutic effects. Marine algal peptides encapsulated in nanocarriers significantly enhance bioavailability, ensuring more efficient absorption and utilization in the body. Preclinical studies have shown promising results, indicating that nanocarrier-based delivery systems can significantly improve the pharmacokinetic profiles and therapeutic outcomes of marine algal peptides. This review delves into the diverse anticancer mechanisms of marine algal peptides, which include inducing apoptosis, disrupting cell cycle progression, and inhibiting angiogenesis. Further research focused on optimizing nanocarrier formulations, conducting comprehensive clinical trials, and continued exploration of marine algal peptides holds great promise for developing innovative, effective, and sustainable cancer therapies.
Collapse
Affiliation(s)
- Visuddho Visuddho
- Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Adi Muradi Muhar
- Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Muhammad Iqhrammullah
- Postgraduate Program of Public Health, Universitas Muhammadiyah Aceh, Banda Aceh 23123, Indonesia
| | - Nelly Mayulu
- Department of Nutrition, Faculty of Health Science, Muhammadiyah Manado University, Manado 95249, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta 11480, Indonesia
| | - Raymond Rubianto Tjandrawinata
- Department of Biotechnology, Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta 12930, Indonesia
| | | | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, Indonesia
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta 55281, Indonesia
| |
Collapse
|
38
|
Zaidi SAA, Chughtai N, Abbassi ZA, Alam J, Malick TS, Salim A, Saleem S. TUSC3, p53 and p21 genetic association with development of oral submucous fibrosis and oral squamous cell carcinoma among addictive tobacco chewers of Pakistan. BMC Oral Health 2024; 24:780. [PMID: 38992585 PMCID: PMC11241966 DOI: 10.1186/s12903-024-04501-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND This study delves into the intricate landscape of oral cancer, a global concern with a high incidence in Asian countries. We focus on oral squamous cell carcinoma (OSCC), primarily driven by the consumption of betel nut and its derivatives. OSCC often arises from premalignant lesions like oral submucous fibrosis (OSF). In Pakistan, OSCC is prevalent among men due to various addictive substances, including smokeless tobacco and chewing materials. Mutations in tumor suppressor genes, such as TP53 and p21, play crucial roles in this malignancy's development. We also explore the involvement of TUSC3 gene deletion in OSCC and OSF. METHODS In this study we investigated demographics, TUSC3 gene expression, deletion analysis, and TP53 and p21 genetic alterations in OSCC and OSF patients (blood and tissue of 50 samples in each condition) who had tobacco derivates usage history. The association analysis was carried out mainly through PCR based genotyping. RESULTS The study's patient cohort (OSCC and OSF) displayed a wide age range from 13 to 65 years (Mean = 32.96 years). Both conditions were more prevalent in males, with a male-female ratio of approximately 2.5:1. Chewing habits analysis revealed high frequencies of gutka use in both OSF and OSCC patients. TUSC3 expression analysis in OSCC cell lines indicated significant downregulation. Genotyping showed no TUSC3 deletion in OSF cases, but a deletion rate of over 22% in OSCC tissue samples. Analysis supported a significant association of TUSC3 deletion with OSCC development but not with OSF. Polymorphism in p53 exon 4 and p21 (rs1801270) were significantly associated with both OSCC and OSF, adding to their pathogenesis. Our findings further revealed a strong correlation between TUSC3 deletion and the excessive use of tobacco and related products, shedding light on the genetic underpinnings of OSCC development. CONCLUSIONS Notably, our study provides a crucial insight into genetic aspects underlying OSCC and OSF in response of addictive consumption of areca nut, betel quid, and tobacco derivatives. A significant association between TUSC3 deletion and OSCC development, along with polymorphisms in TP53 and p21, underscores the importance of further research into the molecular mechanisms driving oral cancer progression for improved diagnosis and treatment outcomes.
Collapse
Affiliation(s)
- Syed Aqib Ali Zaidi
- The Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Pakistan
- Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China
| | | | - Zubair Ahmed Abbassi
- Department of Dentistry, Jinnah Sindh Medical University (JSMU), Karachi, Pakistan
| | - Jehan Alam
- Department of Dentistry, Jinnah Postgraduate Medical Centre (JPMC), Karachi, 75510, Pakistan
| | - Tuba Shakil Malick
- Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, Pakistan
| | - Saima Saleem
- The Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Pakistan.
| |
Collapse
|
39
|
Giarratana AO, Prendergast CM, Salvatore MM, Capaccione KM. TGF-β signaling: critical nexus of fibrogenesis and cancer. J Transl Med 2024; 22:594. [PMID: 38926762 PMCID: PMC11201862 DOI: 10.1186/s12967-024-05411-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
The transforming growth factor-beta (TGF-β) signaling pathway is a vital regulator of cell proliferation, differentiation, apoptosis, and extracellular matrix production. It functions through canonical SMAD-mediated processes and noncanonical pathways involving MAPK cascades, PI3K/AKT, Rho-like GTPases, and NF-κB signaling. This intricate signaling system is finely tuned by interactions between canonical and noncanonical pathways and plays key roles in both physiologic and pathologic conditions including tissue homeostasis, fibrosis, and cancer progression. TGF-β signaling is known to have paradoxical actions. Under normal physiologic conditions, TGF-β signaling promotes cell quiescence and apoptosis, acting as a tumor suppressor. In contrast, in pathological states such as inflammation and cancer, it triggers processes that facilitate cancer progression and tissue remodeling, thus promoting tumor development and fibrosis. Here, we detail the role that TGF-β plays in cancer and fibrosis and highlight the potential for future theranostics targeting this pathway.
Collapse
Affiliation(s)
- Anna O Giarratana
- Northwell Health - Peconic Bay Medical Center, 1 Heroes Way, Riverhead, NY, 11901, USA.
| | | | - Mary M Salvatore
- Department of Radiology, Columbia University, New York, NY, 11032, USA
| | | |
Collapse
|
40
|
Schaufelberger SA, Schaettin M, Azzarito G, Rosselli M, Leeners B, Dubey RK. 2-Methoxyestradiol, an Endogenous 17β-Estradiol Metabolite, Induces Antimitogenic and Apoptotic Actions in Oligodendroglial Precursor Cells and Triggers Endoreduplication via the p53 Pathway. Cells 2024; 13:1086. [PMID: 38994940 PMCID: PMC11240791 DOI: 10.3390/cells13131086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
The abnormal growth of oligodendrocyte precursor cells (OPCs) significantly contributes to the progression of glioblastoma tumors. Hence, molecules that block OPC growth may be of therapeutic importance in treating gliomas. 2-Methoxyestradiol (2ME), an endogenous tubulin-interacting metabolite of estradiol, is effective against multiple proliferative disorders. Based on its anti-carcinogenic and anti-angiogenic actions, it is undergoing phase II clinical trials. We hypothesize that 2ME may prevent glioma growth by targeting OPC growth. Here, we tested this hypothesis by assessing the impact of 2ME on the growth of an OPC line, "Oli-neu", and dissected the underlying mechanism(s). Treatment with 2ME inhibited OPC growth in a concentration-dependent manner, accompanied by significant upregulation in the expression of p21 and p27, which are negative cell-cycle regulators. Moreover, treatment with 2ME altered OPC morphology from multi-arm processes to rounded cells. At concentrations of 1uM and greater, 2ME induced apoptosis, with increased expressions of caspase 3, PARP, and caspase-7 fragments, externalized phosphatidylserine staining/APOPercentage, and increased mitochondrial activity. Flow cytometry and microscopic analysis demonstrated that 2ME triggers endoreduplication in a concentration-dependent fashion. Importantly, 2ME induced cyclin E, JNK1/2, and p53 expression, as well as OPC fusion, which are key mechanisms driving endoreduplication and whole-genome duplication. Importantly, the inhibition of p53 with pifithrin-α rescued 2ME-induced endoreduplication. The pro-apoptotic and endoreduplication actions of 2ME were accompanied by the upregulation of survivin, cyclin A, Cyclin B, Cyclin D2, and ppRB. Similar growth inhibitory, apoptotic, and endoreduplication effects of 2ME were observed in CG4 cells. Taken together, our findings provide evidence that 2ME not only inhibits OPC growth and triggers apoptosis, but also activates OPCs into survival (fight or flight) mode, leading to endoreduplication. This inherent survival characteristic of OPCs may, in part, be responsible for drug resistance in gliomas, as observed for many tubulin-interacting drugs. Importantly, the fate of OPCs after 2ME treatment may depend on the cell-cycle status of individual cells. Combining tubulin-interfering molecules with drugs such as pifithrin-α that inhibit endoreduplication may help inhibit OPC/glioma growth and limit drug resistance.
Collapse
Affiliation(s)
- Sara. A. Schaufelberger
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Martina Schaettin
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Giovanna Azzarito
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Marinella Rosselli
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Brigitte Leeners
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
| | - Raghvendra K. Dubey
- Department of Obstetrics and Gynaecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland (G.A.); (M.R.); (B.L.)
- Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
41
|
Fei Y, Ma M, Gan L, Xu M, Yang Y, Huang D, Sheng W. Clinicopathological significance and prognostic analysis of p21 and EGFR in colorectal cancer: a retrospective analysis on 12 319 cases in China. J Clin Pathol 2024:jcp-2024-209450. [PMID: 38886043 DOI: 10.1136/jcp-2024-209450] [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: 02/16/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024]
Abstract
AIMS Colorectal cancer (CRC) is the third most common malignancy worldwide. Accurate pathological diagnosis and predictive abilities for treatment response and prognosis are crucial for patients with CRC. This study aims to analyse the expressions of p21 and EGFR in CRC and their relationships with clinicopathological characteristics and prognosis to enhance diagnostic and prognostic evaluations. METHODS This study conducted a retrospective analysis of p21 and EGFR expressions in 12 319 Chinese patients with CRC using immunohistochemistry. The relationships between these expressions and clinicopathological characteristics and survival outcomes were explored through statistical and survival analyses. RESULTS Differential expressions of p21 and EGFR in CRC were closely related to clinicopathological characteristics and significantly impacted overall survival (OS). p21 expression was associated with the primary tumour site, mucinous subtype, lymphovascular invasion, perineural invasion, circumferential resection margin, T stage, N stage, tumour, node, metastases (TNM) stage, and mismatch repair status. EGFR expression was related to mucinous subtype, tumour differentiation, lymphovascular invasion, perineural invasion, tumour size, T stage, N stage, TNM stage and BRAF gene mutation. p21 and EGFR expressions were positively correlated (r=0.11). High p21 expression correlated with favourable OS, whereas high EGFR expression predicted poorer OS. A prognostic nomogram incorporating these biomarkers and clinical variables demonstrated robust predictive power for patient survival rates. CONCLUSION p21 and EGFR serve as potential indicators for pathological diagnosis, risk stratification, and predicting treatment efficacy and prognosis in patients with CRC. The study's findings provide valuable references for personalised treatment and prognosis evaluation in clinical practice.
Collapse
Affiliation(s)
- Yang Fei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Mengke Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Lu Gan
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer center, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Zhangjiang Institute, Shanghai, China
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Yu Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Subramaniyam K, Harihar S. An Overview on the Emerging Role of the Plasma Protease Inhibitor Protein ITIH5 as a Metastasis Suppressor. Cell Biochem Biophys 2024; 82:399-409. [PMID: 38355846 DOI: 10.1007/s12013-024-01227-7] [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: 01/30/2023] [Accepted: 02/02/2024] [Indexed: 02/16/2024]
Abstract
Most cancers are not detected until they have progressed to the point of becoming malignant and life-threatening. Chemotherapy and conventional medicines are often ineffective against cancer. Although we have made significant progress, new conceptual discoveries are still required to investigate new treatments. The role of metastasis suppressor genes as a therapeutic option for limiting tumor progression and metastasis has been on the anvil for some time. In this review, we discuss the role of ITIH5 as a metastasis suppressor gene and catalog its involvement in different cancers. We further shed light on the mode of action of ITIH5 based on the available data. The review will provide a new perspective on ITIH5 as an anti-metastatic protein and hopefully serve as an impetus for future studies towards the application of ITIH5 for clinical intervention in targeting metastatic cancers.
Collapse
Affiliation(s)
- Krishnaveni Subramaniyam
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Sitaram Harihar
- Department of Biotechnology, GITAM School of Science, GITAM (Deemed to be) University, Visakhapatnam, 530045, Andhra Pradesh, India.
| |
Collapse
|
43
|
Wang C, Chang R, Li J, Li L. TRIM47 silencing inhibits the malignant biological behaviors of prostate cancer cells by regulating MDM2/p53 signaling. Cell Biochem Biophys 2024; 82:1567-1578. [PMID: 38802602 DOI: 10.1007/s12013-024-01308-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024]
Abstract
Prostate cancer (PCa) is a prevalent male malignancy globally. Tripartite motif 47 (TRIM47) has been reported to be associated with PCa. However, how TRIM47 acts on PCa is still incompletely understood. Here, we explored the biological roles of TRIM47 in PCa cells and investigated its potential regulatory mechanism. TRIM47 expression in PCa cells was detected by qRT-PCR and western blot. After TRIM47 silencing, the viability of PCa cells was measured using CCK-8 method. Flow cytometry was employed to estimate cell cycle. Cell apoptotic level was subjected to appraisement with TUNEL assay. Additionally, wound healing- and transwell assays were adopted for evaluation of migration and invasion of PCa cells. Moreover, the Biogrid database and HDOCK SERVER predicated that TRIM47 could interact with mouse double minute 2 (MDM2), which was detected using the Co-immunoprecipitation (co-IP) assay and glutathione S-transferase (GST) pull-down assay. The expression of proteins in MDM2/p53 signaling was detected by western blot analysis. Results indicated that TRIM47 expression was highly expressed in PCa cells. TRIM47 knockdown inhibited PCa proliferation and cell cycle whereas promoted cell apoptosis. Besides, TRIM47 knockdown significantly inhibited the migration and invasion of PCa cells. In addition, TRIM47 was proved to bind to MDM2 and regulated MDM2/p53 expression. Importantly, MDM2 overexpression counteracted the impacts of TRIM47 knockdown on cell viability, cell cycle, apoptosis, migration and invasion by regulating the MDM2/p53 pathway. Collectively, our results suggested that TRIM47 silencing inhibits the malignant biological behaviors of prostate cancer cells by regulating MDM2/p53 signaling, which may provide a novel therapeutic target for PCa treatment.
Collapse
Affiliation(s)
- Chengyong Wang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China.
| | - Rui Chang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China
| | - Jian Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China
| | - Liqiang Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China
| |
Collapse
|
44
|
Huda F, Hermawan R, Putri T, Dwiwina RG, Berbudi A, Bashari MH. Anticancer Activity of Aaptos suberitoides on Glioblastoma Multiforme Cell Line. Asian Pac J Cancer Prev 2024; 25:1815-1821. [PMID: 38809654 PMCID: PMC11318835 DOI: 10.31557/apjcp.2024.25.5.1815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/18/2024] [Indexed: 05/31/2024] Open
Abstract
OBJECTIVE Glioblastoma Multiforme (GBM) poses a significant challenge due to its high aggressiveness and unfavorable prognosis, with existing treatments demonstrating limited efficacy in prolonging survival rates. This study aimed to assess the anticancer properties of Aaptos suberitoides extracts and fraction on the U87 cell line, serving as a representative model for GBM. METHODS U87 cells were treated with ethanol extracts derived from Aaptos suberitoides, specifically two extracts (OAA-1 and OAA-2) and one ethyl acetate fraction (EA) isolated from specimens collected on Pramuka Island and Tinjil Island. The evaluation encompased microscopic observation and MTT assay to determine the IC50. Subsequently, antiproliferative effects were investigated through apoptosis and cell cycle assays. RESULTS The extract demonstrated cytotoxic activity against U87 cells, with OAA-1 and OAA-2 exhibiting IC50 values of 35.78 μg/mL and 25.38 μg/mL, respectively. OAA-1 notably induced apoptosis at 50 μg/mL and induced cell cycle arrest. On other hand, OAA-2, while also inducing apoptosis significantly, had a lesser impact on cell cycle arrest. In contrast, EA induced significant apoptosis at a concentration of 100 μg/mL. CONCLUSION The ethanol extracts and the ethyl acetate fraction of Aaptos suberitoides emerged as a promising candidate for Glioblastoma Multiforme cancer therapy, showing potential in inhibiting cell proliferation and inducing apoptosis.
Collapse
Affiliation(s)
- Fathul Huda
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia.
| | - Rohim Hermawan
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia.
| | - Tenny Putri
- Laboratory of Advanced Biomedicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia.
| | - Resti Gradia Dwiwina
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia.
| | - Afiat Berbudi
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia.
| | - Muhammad Hasan Bashari
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia.
| |
Collapse
|
45
|
Wang X, Fukumoto T, Noma KI. Therapeutic strategies targeting cellular senescence for cancer and other diseases. J Biochem 2024; 175:525-537. [PMID: 38366629 PMCID: PMC11058315 DOI: 10.1093/jb/mvae015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/12/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024] Open
Abstract
Cellular senescence occurs in response to endogenous or exogenous stresses and is characterized by stable cell cycle arrest, alterations in nuclear morphology and secretion of proinflammatory factors, referred to as the senescence-associated secretory phenotype (SASP). An increase of senescent cells is associated with the development of several types of cancer and aging-related diseases. Therefore, senolytic agents that selectively remove senescent cells may offer opportunities for developing new therapeutic strategies against such cancers and aging-related diseases. This review outlines senescence inducers and the general characteristics of senescent cells. We also discuss the involvement of senescent cells in certain cancers and diseases. Finally, we describe a series of senolytic agents and their utilization in therapeutic strategies.
Collapse
Affiliation(s)
- Xuebing Wang
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo 060-0815, Japan
| | - Takeshi Fukumoto
- Division of Dermatology, Department of Internal Related, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Ken-ichi Noma
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo 060-0815, Japan
- Institute of Molecular Biology, University of Oregon, 1370 Franklin Blvd, Eugene, OR 97403, USA
| |
Collapse
|
46
|
Chae J, Choi Y, Hong J, Kim N, Kim J, Lee HY, Choi J. Anticancer and Antibacterial Properties of Curcumin-Loaded Mannosylated Solid Lipid Nanoparticles for the Treatment of Lung Diseases. ACS APPLIED BIO MATERIALS 2024; 7:2175-2185. [PMID: 38478917 DOI: 10.1021/acsabm.3c01145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Lung cancer and Mycobacterium avium complex infection are lung diseases associated with high incidence and mortality rates. Most conventional anticancer drugs and antibiotics have certain limitations, including high drug resistance rates and adverse effects. Herein, we aimed to synthesize mannose surface-modified solid lipid nanoparticles (SLNs) loaded with curcumin (Man-CUR SLN) for the effective treatment of lung disease. The synthesized Man-CUR SLNs were analyzed using various instrumental techniques for structural and physicochemical characterization. Loading curcumin into SLNs improved the encapsulation efficiency and drug release capacity, as demonstrated by high-performance liquid chromatography analysis. Furthermore, we characterized the anticancer effect of curcumin using the A549 lung cancer cell line. Cells treated with Man-CUR SLN exhibited an increased cellular uptake and cytotoxicity. Moreover, treatment with free CUR could more effectively reduce cancer migration than treatment with Man-CUR SLNs. Similarly, free curcumin elicited a stronger apoptosis-inducing effect than that of Man-CUR SLNs, as demonstrated by reverse transcription-quantitative PCR analysis. Finally, we examined the antibacterial effects of free curcumin and Man-CUR SLNs against Mycobacterium intracellulare (M.i.) and M.i.-infected macrophages, revealing that Man-CUR SLNs exerted the strongest antibacterial effect. Collectively, these findings indicate that mannose-receptor-targeted curcumin delivery using lipid nanoparticles could be effective in treating lung diseases. Accordingly, this drug delivery system can be used to target a variety of cancers and immune cells.
Collapse
Affiliation(s)
- Jayoung Chae
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| | - Joohye Hong
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Namju Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jiwon Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee-Young Lee
- Department of Chemical Engineering, Kumoh National Institute of Technology, 61, Daehak-ro, Gumi-si, Gyeongsangbuk-do 39177, Republic of Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| |
Collapse
|
47
|
Riadi Y, Afzal O, Kumar S, Varadharajan V, Geesi MH. Synthesis of novel ( R)-carvone-tagged thiazolidinone as anticancer leads: characterization, in vitro antiproliferative evaluation and in silico studies. J Biomol Struct Dyn 2024:1-14. [PMID: 38523573 DOI: 10.1080/07391102.2024.2331095] [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: 01/04/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
This work describes the successful synthesis of a series of three novel thiazolidinone-carvone-O-alkyl hybrids through a two-step approach involving heterocyclization and O-alkylation reactions. Comprehensive structural characterization of the obtained products was achieved using NMR and HRMS spectroscopic techniques. This study assessed in vitro antiproliferative activity of synthesized thiazolidinone-carvone-O-alkyl hybrids (5a-c) against various human cancer cell lines, viz. HT-1080 (fibrosarcoma), A-549 (lung cancer), MCF-7 (breast cancer) and MDA-MB-231 (breast cancer). MTT assay revealed promising results for compounds 5b and 5c, demonstrating good antiproliferative activity against A-549 and MCF-7 cell lines comparable to the positive control, Doxorubicin. Compound 5a, harbouring an O-acetoxy group, displayed limited anticancer activity against MCF-7 and MDA-MB-231 cells, with IC50 values of 69.33 ± 0.42 µM and >100 µM, respectively. Docking results confirmed that the compounds 5a-c binds at the active site of p21 with docking scores -2.0, -4.8, and -7.0 kcal/mol, respectively. Compound 5a-c also showed good binding potential against Bcl2 protein with docking score of -4.9, -6.0, -5.5 kcal/mol, respectively. Furthermore, binding energy analysis and dynamics simulation studies of compounds towards p21 and Bcl2 yielded promising results. In PAK4 assay, compound 5c showed comparable potency (IC50 6.76 µM) with the standard control UC2288 (IC50 6.40 µM), while in BCL-2 TR-FRET assay, 5c exhibited good inhibition (IC50 1.78 µM) as compared to Venetoclax (IC50 0.016 µM). In conclusion, compounds 5a-c could be used as a structural framework for the discovery of novel therapeutics to combat different types of cancer.
Collapse
Affiliation(s)
- Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, India
| | | | - Mohammed H Geesi
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
48
|
Xu Y, Zheng Z, Pan H, Zhao M, Zhang J, Peng S, Liu J, Pan W, Yin Z, Xu S, Wei C, Qin JJ, Lin Y, Wan J, Wang M. Kielin/chordin-like protein deficiency aggravates pressure overload-induced cardiac dysfunction and remodeling via P53/P21/CCNB1 signaling in mice. FASEB J 2024; 38:e23513. [PMID: 38421300 DOI: 10.1096/fj.202301841r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/07/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Targeting cardiac remodeling is regarded as a key therapeutic strategy for heart failure. Kielin/chordin-like protein (KCP) is a secretory protein with 18 cysteine-rich domains and associated with kidney and liver fibrosis. However, the relationship between KCP and cardiac remodeling remains unclear. Here, we aimed to investigate the role of KCP in cardiac remodeling induced by pressure overload and explore its potential mechanisms. Left ventricular (LV) KCP expression was measured with real-time quantitative PCR, western blotting, and immunofluorescence staining in pressure overload-induced cardiac remodeling in mice. Cardiac function and remodeling were evaluated in wide-type (WT) mice and KCP knockout (KO) mice by echocardiography, which were further confirmed by histological analysis with hematoxylin and eosin and Masson staining. RNA sequence was performed with LV tissue from WT and KO mice to identify differentially expressed genes and related signaling pathways. Primary cardiac fibroblasts (CFs) were used to validate the regulatory role and potential mechanisms of KCP during fibrosis. KCP was down-regulated in the progression of cardiac remodeling induced by pressure overload, and was mainly expressed in fibroblasts. KCP deficiency significantly aggravated pressure overload-induced cardiac dysfunction and remodeling. RNA sequence revealed that the role of KCP deficiency in cardiac remodeling was associated with cell division, cell cycle, and P53 signaling pathway, while cyclin B1 (CCNB1) was the most significantly up-regulated gene. Further investigation in vivo and in vitro suggested that KCP deficiency promoted the proliferation of CFs via P53/P21/CCNB1 pathway. Taken together, these results suggested that KCP deficiency aggravates cardiac dysfunction and remodeling induced by pressure overload via P53/P21/CCNB1 signaling in mice.
Collapse
Affiliation(s)
- Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
49
|
Cherian E, Goyal M, Mittal N, Mathews S, Sagir M. Elucidating the Anticancer Mechanisms of Cinnamoyl Sulfonamide Hydroxamate: Insights From DNA Content Analysis and Gene Expression Profiling in Squamous Cell Carcinoma. Cureus 2024; 16:e57236. [PMID: 38686281 PMCID: PMC11056768 DOI: 10.7759/cureus.57236] [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] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Oral cancer is a major public health concern worldwide, with oral squamous cell carcinoma (OSCC) being one of its most common subtypes. Despite advances in diagnosis and management of this disease, there remains a need to develop new therapeutic approaches for better outcomes. OBJECTIVE This study aimed to investigate the molecular mechanisms through which cinnamoyl sulfonamide hydroxamate derivatives exert their anticancer effects on OSCC. MATERIALS AND METHODS The derivatives were synthesized via multi-step processes and then characterized at the molecular level. Flow cytometry assay for DNA content and cell cycle distribution, anisidine/toluidine double staining for apoptosis detection, as well as reverse transcription polymerase chain reaction (RT-PCR) gene expression analysis, were performed on OSCC cell lines exposed to cinnamoyl sulfonamide hydroxamate derivatives. RESULTS Flow cytometry unveiled remarkable changes in the distribution of cells throughout the OSCC cell line upon treatment with cinnamoyl sulfonamide hydroxamate derivatives. Consequently, it led to a noticeable decrease in cells at the G0/G1 phase, together with an increase at the S phase, thereby indicating a retardation at various points of the cycle. In addition, apoptotic morphological alterations have been observed by anisidine/toluidine double staining after some treatments with the compounds. RT-PCR analysis showed a marked increase in p21 gene expression levels, further supporting the compounds' ability to induce cell cycle arrest and apoptosis. CONCLUSION The research highlighted the potential of cinnamoyl sulfonamide hydroxamate derivatives as candidates for oral cancer, particularly OSCC treatment, shedding light on their operation at the molecular level and paving the way for the development of targeted therapies that could aid in the cure of oral cancer.
Collapse
Affiliation(s)
- Eapen Cherian
- Oral and Maxillofacial Pathology, Travancore Dental College Medicity, Kollam, IND
| | - Manoj Goyal
- Oral and Maxillofacial Surgery, Santosh Deemed to Be University, Ghaziabad, IND
| | - Neeti Mittal
- Pediatric Dentistry, Santosh Deemed to Be University, Ghaziabad, IND
| | - Susan Mathews
- Prosthodontics, Travancore Dental College, Kollam, IND
| | - Muhammad Sagir
- Conservative Dentistry and Endodontics, Royal Dental College Medicity, Pallakad, IND
| |
Collapse
|
50
|
Cai HQ, Zhang LY, Fu LM, Xu B, Jiao Y. Mutational landscape of TP53 and CDH1 in gastric cancer. World J Gastrointest Surg 2024; 16:276-283. [PMID: 38463349 PMCID: PMC10921187 DOI: 10.4240/wjgs.v16.i2.276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/26/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
In this editorial we comment on an article published in a recent issue of the World J Gastrointest Surg. A common gene mutation in gastric cancer (GC) is the TP53 mutation. As a tumor suppressor gene, TP53 is implicated in more than half of all tumor occurrences. TP53 gene mutations in GC tissue may be related with clinical pathological aspects. The TP53 mutation arose late in the progression of GC and aided in the final switch to malignancy. CDH1 encodes E-cadherin, which is involved in cell-to-cell adhesion, epithelial structure maintenance, cell polarity, differentiation, and intracellular signaling pathway modulation. CDH1 mutations and functional loss can result in diffuse GC, and CDH1 mutations can serve as independent prognostic indicators for poor prognosis. GC patients can benefit from genetic counseling and testing for CDH1 mutations. Demethylation therapy may assist to postpone the onset and progression of GC. The investigation of TP53 and CDH1 gene mutations in GC allows for the investigation of the relationship between these two gene mutations, as well as providing some basis for evaluating the prognosis of GC patients.
Collapse
Affiliation(s)
- Hong-Qiao Cai
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Li-Yue Zhang
- Department of Critical Care Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Li-Ming Fu
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Bin Xu
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|