1
|
Pereira JC, de Sousa RWR, Conceição MLP, do Nascimento MLLB, de Almeida ATA, Dos Reis AC, de Sousa Cavalcante ML, Dos Reis Oliveira C, Martins IRR, Torres-Leal FL, Dittz D, de Castro E Sousa JM, Ferreira PMP, Carneiro da Silva FC. Buthionine sulfoximine acts synergistically with doxorubicin as a sensitizer molecule on different tumor cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:409-431. [PMID: 39815616 DOI: 10.1080/15287394.2024.2448663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The chemotherapeutic drug doxorubicin (DOX) has been widely used for treating solid tumors attributed to its antiproliferative effectiveness; however, its clinical use is limited due to side effects, including cardiotoxicity, myelosuppression, and drug resistance. Combining DOX with buthionine sulfoximine (BSO), a glutathione (GSH) synthesis inhibitor, showed promising results in overcoming these adverse effects, potentially reducing the required DOX dose while maintaining efficacy. The aim of the present study was to examine the effects of different concentrations of BSO and DOX, both individually and in combination, utilizing B16/F10 (murine melanoma), SNB-19 (human glioblastoma), S180 (murine sarcoma), and SVEC4-10 (murine endothelial) cell lines. Cell viability, migration, and clonogenicity were assessed using the following assays MTT, scratch, and colony formation. Antioxidant levels of GSH, as well as activities catalase (CAT), and superoxide dismutase (SOD) were measured. BSO alone exhibited minimal cytotoxic effects, while DOX alone reduced cell viability significantly. The combination of BSO+DOX decreased IC50 values for most cell lines, demonstrating a synergistic effect, especially in B16/F10, S180, and SVEC4-10 cells. BSO+DOX combination significantly inhibited cell migration and clonogenicity compared to DOX alone. While GSH levels were decreased with BSO+DOX treatment activities of CAT and SOD increased following DOX administration but remained unchanged by BSO. These results suggest that BSO may be considered a valuable tool to improve DOX therapeutic efficacy, particularly in cases of chemotherapy-resistant tumors, as BSO enhances DOX activity while potentially reducing systemic chemotherapeutic drug toxicity.
Collapse
Affiliation(s)
- Joedna Cavalcante Pereira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Rayran Walter Ramos de Sousa
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Micaely Lorrana Pereira Conceição
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | | | - Ana Tárcila Alves de Almeida
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Antonielly Campinho Dos Reis
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| | - Mickael Laudrup de Sousa Cavalcante
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Camila Dos Reis Oliveira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Italo Rossi Roseno Martins
- Academic Unit of Life Sciences, Teachers' Forming Center, Federal University of Campina Grande, Cajazeiras-PB, Brazil
| | - Francisco Leonardo Torres-Leal
- Metabolic Diseases, Exercise and Nutrition Research Group (Domen), Laboratory of Metabolic Diseases Glauto Tuquarre, Department of Biophysics and Physiology, Federal University of Piaui, Teresina-PI, Brazil
| | - Dalton Dittz
- Laboratory of Antineoplastic Pharmacology (Lafan), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| | - João Marcelo de Castro E Sousa
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
| | - Felipe Cavalcanti Carneiro da Silva
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina-PI, Brazil
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina-PI, Brazil
| |
Collapse
|
2
|
Zeng Y, Tao Y, Du G, Huang T, Chen S, Fan L, Zhang N. Advances in the mechanisms of HIF-1α-enhanced tumor glycolysis and its relation to dedifferentiation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025:S0079-6107(25)00024-0. [PMID: 40373959 DOI: 10.1016/j.pbiomolbio.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Metabolic reprogramming, a hallmark of malignancy, enables tumor cells to adapt to the harsh and dynamic tumor microenvironment (TME) by altering metabolic pathways. Hypoxia, prevalent in solid tumors, activates hypoxia inducible factor 1α (HIF-1α). HIF-1α drives metabolic reprogramming, enhancing glycolysis primarily through the Warburg effect to reduce oxygen dependence and facilitate tumor cell growth/proliferation. The above process is associated with accelerated tumor cell dedifferentiation and enhanced stemness, generating cancer stem cells (CSCs) which possesses the potential for self-renewal and differentiation that can differentiate into a wide range of subtypes of tumor cells and fuel tumor heterogeneity, metastasis, and recurrence, complicating therapy. This review examines the HIF-1α-glycolysis-dedifferentiation crosstalk mechanisms, expecting that indirect inhibition of HIF-1α by targeting metabolic enzymes, metabolites, or their signaling pathways will offer an effective therapeutic strategy to improve the cancer treatment outcomes.
Collapse
Affiliation(s)
- Yu Zeng
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Yonggang Tao
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Guotu Du
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Tianyu Huang
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Shicheng Chen
- Zunyi China, The Second Affiliated Hospital of Zunyi Medical University(2)
| | - Longmei Fan
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Neng Zhang
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1).
| |
Collapse
|
3
|
Vinogradov AE, Anatskaya OV. "Cell dedifferentiation" versus "evolutionary reversal" theories of cancer: The direct contest of transcriptomic features. Int J Cancer 2025; 156:1802-1813. [PMID: 39888036 DOI: 10.1002/ijc.35352] [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: 10/11/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Cell dedifferentiation is considered an important hallmark of cancer. The atavistic reversal to a unicellular-like (UC) state is a less widely accepted concept, so far not included in the conventional hallmarks. The activated expression of ontogenetically earlier and evolutionary earlier genes in cancers supports both theories because ontogenesis partially recapitulates phylogenesis during cell differentiation (the cellular biogenetic law). We directly contested both types of gene signatures in stem vs. differentiated and cancer vs. normal cells, using meta-analysis of human single-cell transcriptomes (totally, 38 pairwise comparisons involving over 18,600 cells). Because compared cells can differ in proliferation rate, the correction for cell cycle activity was applied. Taken together as multiple variables in stem vs. differentiated cells analyses, the ontogenetic signature excluded the UC signature from predictive variables, usually even forcing it to change the sign of prediction (from plus to minus). In contrast, in cancer vs. normal cells, the UC signature excluded the ontogenetic signature. Thus, the direct contest decided in favor of the atavistic theory and placed a UC-like state as a central hallmark of cancer, which has a plausible evolutionarily formed mechanism (UC attractor) and can generate other hallmarks. These data suggest a paradigm shift in the understanding of oncogenesis and propose an integrative framework for cancer research. In a practical sense, the upregulation of UC signature over ontogenetic signature indicates potential tumorigenicity, which can be used in early diagnostics and regenerative medicine. For therapy, these results suggest the UC center of cellular networks as a universal target.
Collapse
Affiliation(s)
| | - Olga V Anatskaya
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
4
|
Mieland AO, Petrosino G, Dejung M, Chen JX, Fulzele A, Mahmoudi F, Tu JW, Mustafa AHM, Zeyn Y, Hieber C, Bros M, Schnöder TM, Heidel FH, Najafi S, Oehme I, Hofmann I, Schutkowski M, Hilscher S, Kosan C, Butter F, Bhatia S, Sippl W, Krämer OH. The protein deacetylase HDAC10 controls DNA replication in malignant lymphoid cells. Leukemia 2025:10.1038/s41375-025-02612-8. [PMID: 40301616 DOI: 10.1038/s41375-025-02612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025]
Abstract
Histone deacetylases (HDACs) comprise a family of 18 epigenetic modifiers. The biologically relevant functions of HDAC10 in leukemia cells are enigmatic. We demonstrate that human cultured and primary acute B cell/T cell leukemia and lymphoma cells require the catalytic activity of HDAC10 for their survival. In such cells, HDAC10 controls a MYC-dependent transcriptional induction of the DNA polymerase subunit POLD1. Consequently, pharmacological inhibition of HDAC10 causes DNA breaks and an accumulation of poly-ADP-ribose chains. These processes culminate in caspase-dependent apoptosis. PZ48 does not damage resting and proliferating human normal blood cells. The in vivo activity of PZ48 against ALL cells is verified in a Danio rerio model. These data reveal a nuclear function for HDAC10. HDAC10 controls the MYC-POLD1 axis to maintain the processivity of DNA replication and genome integrity. This mechanistically defined "HDAC10ness" may be exploited as treatment option for lymphoid malignancies.
Collapse
Affiliation(s)
- Andreas O Mieland
- Institute of Toxicology, Mainz University Medical Center, Mainz, Germany
| | - Giuseppe Petrosino
- Institute of Molecular Biology (IMB), Core Facility Bioinformatics, Mainz, Germany
| | - Mario Dejung
- Institute of Molecular Biology (IMB), Core Facility Proteomics, Mainz, Germany
| | - Jia-Xuan Chen
- Institute of Molecular Biology (IMB), Core Facility Proteomics, Mainz, Germany
| | - Amitkumar Fulzele
- Institute of Molecular Biology (IMB), Core Facility Proteomics, Mainz, Germany
| | - Fereshteh Mahmoudi
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Jia-Wey Tu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany
| | - Al-Hassan M Mustafa
- Institute of Toxicology, Mainz University Medical Center, Mainz, Germany
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Yanira Zeyn
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | - Christoph Hieber
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | - Tina M Schnöder
- Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover MedicalSchool (MHH), Hannover, Germany
| | - Florian H Heidel
- Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover MedicalSchool (MHH), Hannover, Germany
- Leibniz Institute on Aging, Fritz-Lipmann-Institute, Jena, Germany
| | - Sara Najafi
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany
- National Center for Tumor Diseases Heidelberg, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany
- National Center for Tumor Diseases Heidelberg, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Ilse Hofmann
- Core Facility Antibodies, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mike Schutkowski
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sebastian Hilscher
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christian Kosan
- Friedrich-Schiller-University Jena, Faculty of Biological Sciences Center for Molecular Biomedicine (CMB) Department of Biochemistry Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Falk Butter
- Institute for Molecular Virology and Cell Biology (IMVZ), Greifswald, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120, Halle (Saale), Germany.
| | - Oliver H Krämer
- Institute of Toxicology, Mainz University Medical Center, Mainz, Germany.
| |
Collapse
|
5
|
Sonar PV, Singh AK, Mandadi S, Sharma NK. Expanding horizons of cancer immunotherapy: hopes and hurdles. Front Oncol 2025; 15:1511560. [PMID: 40352591 PMCID: PMC12061710 DOI: 10.3389/fonc.2025.1511560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Tumor displays various forms of tumor heterogeneity including immune heterogeneity that allow cancer cells to survive during conventional anticancer drug interventions. Thus, there is a strong rationale for overcoming anticancer drug resistance by employing the components of immune cells. Using the immune system to target tumor cells has revolutionized treatment. Recently, significant progress has been achieved at preclinical and clinical levels to benefit cancer patients. Approach A review of literature from the past ten years across PubMed, Scopus, and Web of Science focused on immunotherapy strategies. These include immune checkpoint inhibitors (ICIs), tumor-infiltrating lymphocyte therapy, antibody-drug conjugates (ADCs), cancer vaccines, CAR T-cell therapy, and the role of the gut microbiome. Conclusion While immunotherapy outcomes have improved, particularly for tumor types such as melanoma and non-small cell lung cancer (NSCLC), challenges persist regarding predictive biomarker identification and better management. Ongoing research on modifiers of immune function like gut microbiome-derived metabolites, next-generation ADCs, and new classes of biologics is warranted. Overall, continued investigation toward optimizing synergistic immunotherapeutic combinations through strategic drug delivery systems is imperative for preclinical and clinical success in cancer patients.
Collapse
Affiliation(s)
- Priyanka Vijay Sonar
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Anuj Kumar Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Sravan Mandadi
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
6
|
Ghasemi M, Nowroozzadeh MH, Ghorat F, Iraji A, Hashempur MH. Piperine and its nanoformulations: A mechanistic review of their anti-cancer activities. Biomed Pharmacother 2025; 187:118075. [PMID: 40273688 DOI: 10.1016/j.biopha.2025.118075] [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: 12/14/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Piperine, an active compound found in black pepper, exhibits promising anti-cancer properties by targeting critical signaling pathways involved in cancer cell proliferation, migration, and invasion. This review explores the diverse mechanisms through which piperine exerts its effects, including inhibition of the PI3K/Akt/mTOR and ERK1/2 pathways, activation of p38 and JNK pathways, and suppression of NF-kB/AP-1 signaling. Piperine disrupts Wnt/β-catenin signaling by inhibiting β-catenin nuclear translocation and TCF binding, thereby impairing cancer cell growth and metastasis. Additionally, piperine demonstrates anti-inflammatory actions by reducing CXCL8 expression and modulating the p38 MAPK and JNK pathways. To overcome the issues of low solubility and bioavailability, several nanoformulations of piperinewere developed, such as polymer nanoparticles, nanoemulsion, liposomes, micelles, metal-organic frameworks and inorganic carriers, establishing promising cytotoxicity, prolonged-release, enhanced cellular influx, and directed drug delivery. The mechanisms involve G₀ and G₂/M arrest of the cell cycle, mitochondria-mediated apoptosis (involving Bax/Bcl-2 modulation and caspase activation), and cancer celldeath. In vivo studies underscore the efficacy of piperine, while synergistic effects with other natural products and chemotherapy highlight its potential as a versatile therapeutic agent as an anticancer agent. These findings underscore piperine's potential as a multifaceted therapeutic agent for cancer treatment, emphasizing its diverse mechanisms of action and promising role in oncology.
Collapse
Affiliation(s)
- Mahshad Ghasemi
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - M Hossein Nowroozzadeh
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Ghorat
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Nakashima M, Fukumoto A, Matsuda S. Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma. Diseases 2025; 13:111. [PMID: 40277821 PMCID: PMC12025462 DOI: 10.3390/diseases13040111] [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: 01/12/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant form of primary liver cancer. Intricate networks linked to the host immune system may be associated with the pathogenesis of HCC. A huge amount of interdisciplinary medical information for the treatment of HCC has been accumulated over recent years. For example, advances in new immunotherapy have improved the results of treatment for HCC. This approach can be advantageously combined with standard conventional treatments such as surgical resection to improve the therapeutic effect. However, several toxic effects of treatments may pose a significant threat to human health. Now, a shift in mindset is important for achieving superior cancer therapy, where probiotic therapy may be considered, at least within the bounds of safety. The interplay between the gut microbiota and immune system could affect the efficacy of several anticancer treatments, including of immune checkpoint therapy via the alteration of Th17 cell function against various malignant tumors. Here, some recent anticancer techniques are discussed, whereby the growth of HCC may be effectively and safely repressed by probiotic therapy.
Collapse
Affiliation(s)
| | | | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
8
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
9
|
Cai J, He M, Wang Y, Zhang H, Xu Y, Wang Y, You C, Gao H. Discovery of a novel microtubule destabilizing agent targeting the colchicine site based on molecular docking. Biochem Pharmacol 2025; 234:116804. [PMID: 39956210 DOI: 10.1016/j.bcp.2025.116804] [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: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Although a number of microtubule-targeting agents have been used in tumour therapy, their resistance and drug toxicity pose clinical challenges. Microtubule destabilizing agents (MDAs) targeting the colchicine site were found to have the advantage of being able to overcome drug resistance. In our previous studies, we identified a novel MDA from the compound database based on virtual screening methods. Its chemical formula is C23H19N3O5S, abbreviated as C10. In this study, molecular docking methods confirmed that the binding pattern of C10 to tubulin is similar to that of colchicine. Immunofluorescence staining and tubulin polymerization experiments showed that C10 disrupts the microtubule network and reduces the polymerization efficiency of tubulin. Cell proliferation and toxicity assay showed that C10 could effectively inhibit the growth of tumour cells. After 72 h treatment, the semi-inhibitory concentrations of A549, MCF-7 and HepG2 were 18.83 μM, 16.32 μM and 16.92 μM. Colony formation assay and EdU staining also showed that C10 significantly inhibited the proliferative capacity of tumour cells. Meanwhile, it was found by wound healing and transwell assay that the migration and invasive ability of tumour cells were relatively weakened after treatment with C10. Furthermore, flow cytometry analysis and western blot revealed that C10 reduced the expression of the B-cell lymphoma 2 (BCL-2) and upregulated the level of the Bcl-2-associated X protein (BAX), which in turn activated caspase-3 to promote apoptosis. Finally, the vivo studies in animal showed that C10 significantly inhibited the growth of tumour in nude mice without significant drug toxicity. Thus, C10 may be a colchicine binding site inhibitor with anticancer potential. Abbreviations: BAX, bcl-2-associated X protein; BCL-2, the B-cell lymphoma 2; BSA, bovine albumin; CBSIs, colchicine binding site inhibitors; CCK-8, cell counting kits-8; DAPI, 4',6-diamidino-2-phenylindole; DMSO, dimethyl sulfoxide; EdU, 5-ethynyl-2'-deoxyuridine; FITC, fluorescein Isothiocyanate; GAPDH,glyceraldehyde-3-phosphate dehydrogenase; H-E, hematoxylin-eosin; HRP, horseradish Peroxidase; IHC, immunohistochemistry; MDAs, microtubule destabilizing agents; MSAs, microtubule stabilizing agents; MTAs, microtubule-targeting agents; PBS, phosphate buffered saline; PI, propidium Iodide; PVDF, polyvinylidenefluoride.
Collapse
Affiliation(s)
- Jiangying Cai
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, PR China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730070, PR China
| | - Miao He
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, PR China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730070, PR China
| | - Yingying Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, PR China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730070, PR China
| | - Hui Zhang
- College of Life Science, Northwest Normal University, Lanzhou 730070, PR China
| | - Yaxin Xu
- Laboratory Medicine Center, Gansu Provincial Hospital, Lanzhou 730000, PR China
| | - Yubin Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, PR China
| | - Chongge You
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, PR China.
| | - Hongwei Gao
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, PR China.
| |
Collapse
|
10
|
Zhang M, Wei J, Sun Y, He C, Ma S, Pan X, Zhu X. The efferocytosis process in aging: Supporting evidence, mechanisms, and therapeutic prospects for age-related diseases. J Adv Res 2025; 69:31-49. [PMID: 38499245 PMCID: PMC11954809 DOI: 10.1016/j.jare.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Aging is characterized by an ongoing struggle between the buildup of damage caused by a combination of external and internal factors. Aging has different effects on phagocytes, including impaired efferocytosis. A deficiency in efferocytosis can cause chronic inflammation, aging, and several other clinical disorders. AIM OF REVIEW Our review underscores the possible feasibility and extensive scope of employing dual targets in various age-related diseases to reduce the occurrence and progression of age-related diseases, ultimately fostering healthy aging and increasing lifespan. Key scientific concepts of review Hence, the concurrent implementation of strategies aimed at augmenting efferocytic mechanisms and anti-aging treatments has the potential to serve as a potent intervention for extending the duration of a healthy lifespan. In this review, we comprehensively discuss the concept and physiological effects of efferocytosis. Subsequently, we investigated the association between efferocytosis and the hallmarks of aging. Finally, we discuss growing evidence regarding therapeutic interventions for age-related disorders, focusing on the physiological processes of aging and efferocytosis.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
11
|
Lin X, Zhang J, Liang J, Ji D, Huang ZS, Li D. Selective Up-Regulation of Tumor Suppressor Gene Retinoblastoma by Bisacridine Derivative Through Gene Promoter Quadruplex Structures for Cancer Treatment. Int J Mol Sci 2025; 26:1417. [PMID: 40003883 PMCID: PMC11855212 DOI: 10.3390/ijms26041417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
The retinoblastoma (RB) gene is an important tumor suppressor gene with a higher mutation frequency than other tumor suppressor genes. The mutation or inactivation of RB has been found in various cancers. The discovery of small molecules to promote RB expression is an effective anti-cancer strategy. Special DNA secondary structures with G-quadruplex and i-motif on the RB promoter could act as "molecular switches" for gene transcriptional regulation and are potentially important targets for the development of new anti-cancer drugs. After extensive screening, we found that the bisacridine derivative A06 had selective binding and destabilization for both the G-quadruplex and i-motif on the RB promoter, which significantly up-regulated RB gene transcription and translation, resulting in the inhibition of tumor cell proliferation and metastasis. A06 exhibited potent anti-tumor activity on Hela cells and strongly suppressed tumor growth on the Hela xenograft mice model without significant toxicity. In comparison, A02 exhibited strong binding and destabilization to the RB promoter G-quadruplex only, which showed a much weaker effect than A06 on regulating RB expression and producing anti-tumor activity. As we know, this is the first study for up-regulating a tumor suppressor gene through destabilization of both the G-quadruplex and i-motif on the gene promoter, which provides a new strategy for innovative anti-cancer drug discovery and development.
Collapse
Affiliation(s)
| | | | | | | | | | - Ding Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou University City, Guangzhou 510006, China; (X.L.); (J.Z.); (J.L.); (D.J.); (Z.-S.H.)
| |
Collapse
|
12
|
Rauf A, Olatunde A, Akram Z, Hemeg HA, Aljohani ASM, Al Abdulmonem W, Khalid A, Khalil AA, Islam MR, Thiruvengadam R, Kim S, Thiruvengadam M. The Role of Pomegranate ( Punica granatum) in Cancer Prevention and Treatment: Modulating Signaling Pathways From Inflammation to Metastasis. Food Sci Nutr 2025; 13:e4674. [PMID: 39898127 PMCID: PMC11782917 DOI: 10.1002/fsn3.4674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 02/04/2025] Open
Abstract
Punica granatum, commonly known as pomegranate, is a traditional medicinal agent owing to its antiquity. The scientific literature has shown that pomegranate extracts exhibit favorable modulation of diverse signaling pathways. These pathways encompass those implicated in inflammation, angiogenesis, hyperproliferation, cellular transformation, tumorigenesis initiation, and ultimately, a reduction in advanced metastasis and tumorigenesis. Pomegranate extracts in this context can be attributed to their high polyphenol content, which has been observed to possess inhibitory properties toward specific signaling pathways associated with cancer. As a formidable pathology, cancer is the most significant cause of death worldwide after cardiovascular disease. The annual incidence of cancer-related mortality has increased progressively. Modifying one's dietary patterns, engaging in regular physical exercise, and maintaining an optimal body mass index are three straightforward measures that an individual may undertake to mitigate their susceptibility to cancer. Incorporating diverse vegetables and fruits into one's dietary regimen exhibits promising potential for preventing a minimum of 20% cancer incidence and approximately 200,000 cancer-related mortalities annually. Vegetables and fruits contain high levels of minerals and phytochemicals, which help alleviate and prevent the harmful effects of cancer. These substances are safe and exhibit minimal toxicity in biological systems. Furthermore, they exhibit antioxidant properties and have garnered extensive approval for their use as nutritional supplements. Pomegranates are used in ancient cultures to prevent and treat various diseases. Extensive research on pomegranate extract, fruit, oil, and juice has revealed promising findings regarding their potential anti-proliferative, anti-tumorigenic, and anti-inflammatory properties through the modification of various signaling pathways related to cancer, thus demonstrating their potential as drugs to prevent and treat cancer. Emerging research indicates that pomegranate can potentially prevent and treat different cancers, including prostate, bladder, breast, skin, lung, and colon cancer.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of ChemistryUniversity of SwabiAnbarKhyber PakhtunkhwaPakistan
| | - Ahmed Olatunde
- Department of Medical BiochemistryAbubakar Tafawa Balewa UniversityBauchiNigeria
| | - Zuneera Akram
- Department of Pharmacology, Faculty of Pharmaceutical SciencesBaqai Medical UniversityKarachiPakistan
| | - Hassan A. Hemeg
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesTaibah UniversityAl‐Medinah, Al‐MonawaraSaudi Arabia
| | - Abdullah S. M. Aljohani
- Department of Medical Biosciences, College of Veterinary MedicineQassim UniversityBuraydahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraydahSaudi Arabia
| | - Ahood Khalid
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahorePunjabPakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahorePunjabPakistan
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health SciencesDaffodil International UniversityDhakaBangladesh
| | - Rekha Thiruvengadam
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences (SIMATS)Saveetha UniversityChennaiIndia
| | - Seung‐Hyun Kim
- Department of Crop Science, College of Sanghuh Life ScienceKonkuk UniversitySeoulRepublic of Korea
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life ScienceKonkuk UniversitySeoulRepublic of Korea
| |
Collapse
|
13
|
Yao K, Fan H, Yang T, Yang C, Wang G, Li X, Ji XY, Wang Q, Lv S, Guo S. Identification of MYC and STAT3 for early diagnosis based on the long noncoding RNA-mRNA network and bioinformatics in colorectal cancer. Front Immunol 2025; 15:1497919. [PMID: 39830506 PMCID: PMC11739134 DOI: 10.3389/fimmu.2024.1497919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Background Colorectal cancer (CRC) ranks among the top three cancers globally in both incidence and mortality, posing a significant public health challenge. Most CRC cases are diagnosed at intermediate to advanced stages, and reliable biomarkers for early detection are lacking. Long non-coding RNAs (lncRNAs) have been implicated in various cancers, including CRC, playing key roles in tumor development, progression, and prognosis. Methods A comprehensive search of the PubMed database was conducted to identify relevant studies on the early diagnosis of CRC. Bioinformatics analysis was performed to explore lncRNA-mRNA networks, leading to the identification of five potential blood biomarkers. Expression analysis was carried out using the GEPIA and GEO online databases, focusing on MYC and STAT3. Differential expression between normal and CRC tissues was assessed, followed by Receiver Operating Characteristic (ROC) analysis to evaluate the diagnostic potential of these markers. Quantitative Real-Time PCR (qRT-PCR) was performed to validate MYC and STAT3 expression levels, and findings were further confirmed using the Human Protein Atlas (HPA) database. Results Database analysis revealed significant differential expression of MYC and STAT3 between normal and CRC tissues. ROC analysis demonstrated the diagnostic potential of these markers. qRT-PCR validation confirmed the differential expression patterns observed in the databases. Validation through the HPA database further supported these findings, confirming the potential of MYC and STAT3 as diagnostic biomarkers for CRC. Conclusion Our results suggest that MYC and STAT3 are promising diagnostic biomarkers for CRC, offering new insights into its pathophysiology and potential for targeted therapies.
Collapse
Affiliation(s)
- Kunhou Yao
- Department of General Surgery, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Hao Fan
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Tiancheng Yang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Can Yang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Guibin Wang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Xingwang Li
- Department of General Surgery, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Xin-Ying Ji
- Department of General Surgery, Huaxian County People’s Hospital, Huaxian, Henan, China
| | - Qun Wang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Shaojiang Lv
- Department of General Surgery, Huaxian County People’s Hospital, Huaxian, Henan, China
| | - Shihao Guo
- Department of Colorectal Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Bidram M, Ganjalikhany MR. Bioactive peptides from food science to pharmaceutical industries: Their mechanism of action, potential role in cancer treatment and available resources. Heliyon 2024; 10:e40563. [PMID: 39654719 PMCID: PMC11626046 DOI: 10.1016/j.heliyon.2024.e40563] [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: 02/08/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer is known as the main cause of mortality in the world, and every year, the rate of incidence and death due to cancer is increasing. Bioactive peptides are one of the novel therapeutic options that are considered a suitable alternative to toxic chemotherapy drugs because they limit side effects with their specific function. In fact, bioactive peptides are short amino acid sequences that obtain diverse physiological functions to maintain human health after being released from parent proteins. This group of biological molecules that can be isolated from different types of natural protein sources has attracted much attention in the field of pharmaceutical and functional foods production. The current article describes the therapeutic benefits of bioactive peptides and specifically and extensively reviews their role in cancer treatment, available sources for discovering anticancer peptides, mechanisms of action, production methods, and existing challenges.
Collapse
Affiliation(s)
- Maryam Bidram
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohamad Reza Ganjalikhany
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
15
|
Pasupalak JK, Rajput P, Gupta GL. Gut microbiota and Alzheimer's disease: Exploring natural product intervention and the Gut-Brain axis for therapeutic strategies. Eur J Pharmacol 2024; 984:177022. [PMID: 39362390 DOI: 10.1016/j.ejphar.2024.177022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Numerous studies conducted over the last ten years have shown a strong correlation between the gut microbiota and the onset and progression of Alzheimer's disease (AD). However, the exact underlying mechanism is still unknown. An ongoing communication mechanism linking the gut and the brain is highlighted by the term "microbiota-gut-brain axis," which was originally coined the "gut-brain axis." Key metabolic, endocrine, neurological, and immunological mechanisms are involved in the microbiota‒gut‒brain axis and are essential for preserving brain homeostasis. Thus, the main emphasis of this review is how the gut microbiota contributes to the development of AD and how various natural products intervene in this disease. The first part of the review provides an outline of various pathways and relationships between the brain and gut microbiota, and the second part provides various mechanisms involved in the gut microbiota and AD. Finally, this review provides knowledge about natural products and their effectiveness in treating gut microbiota-induced AD. AD may be treated in the future by altering the gut microbiota with a customized diet, probiotics/prebiotics, plant products, and natural products. This entails altering the microbiological partners and products (such as amyloid protein) that these partners generate.
Collapse
Affiliation(s)
- Jajati K Pasupalak
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Prabha Rajput
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Girdhari Lal Gupta
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India.
| |
Collapse
|
16
|
Turk A, Metin TO, Kuloglu T, Yilmaz M, Artas G, Ozercan IH, Hancer S. Isthmin-1 and spexin as promising novel biomarker candidates for invasive ductal breast carcinoma. Tissue Cell 2024; 91:102601. [PMID: 39520846 DOI: 10.1016/j.tice.2024.102601] [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: 09/17/2023] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Breast cancer is one of the most common malignant tumors and a leading cause of cancer-related death in women. Research is focusing on biomarkers linked to breast cancer, particularly two novel proteins: isthmin-1 (ISM-1) and spexin (SPX), which require further investigation. MATERIAL AND METHODS The study involved 20 healthy controls and 60 patients with invasive ductal carcinoma, categorized into three groups: Grade I (n=20), Grade II (n=20), and Grade III (n=20). Levels of ISM-1 and SPX in tissue were analyzed using immunohistochemistry alongside the clinicopathologic data of patients. RESULTS There were no statistically significant differences in age, menopausal status, ER, PR, and Cerb-B2 values across grades (p>0.05). Tumor diameters showed a significant increase in Grade I compared to Grade II (p<0.05), while no significant difference was noted between Grade II and Grade III, although diameters were larger in Grade III compared to Grade I (p<0.05). Notably, ISM-1 immunoreactivity decreased, and SPX immunoreactivity increased significantly across all grades compared to normal tissue (p<0.05), with no significant differences between tumor grades for these markers (p>0.05). CONCLUSIONS This study presents new findings on ISM-1 and SPX expression in invasive ductal breast carcinoma. The decrease in ISM-1 and increase in SPX suggest a need for further research into the relationship between adipokines and tumor development in breast cancer.
Collapse
Affiliation(s)
- Ahmet Turk
- Department of Histology and Embryology, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey.
| | - Tuba Ozcan Metin
- Department of Histology and Embryology, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Tuncay Kuloglu
- Department of Histology and Embryology, Faculty of Medicine, Firat University, Elazıg, Turkey
| | - Mustafa Yilmaz
- Department of Emergency Medicine, Firat University School of Medicine, Elazig, Turkey
| | - Gokhan Artas
- Department of Pathology, Firat University, School of Medicine, Elazig, Turkey
| | - I Hanifi Ozercan
- Department of Pathology, Firat University, School of Medicine, Elazig, Turkey
| | - Serhat Hancer
- Department of Histology and Embryology, Faculty of Medicine, Firat University, Elazıg, Turkey
| |
Collapse
|
17
|
Llamoza-Torres CJ, Fuentes-Pardo M, Ramos-Molina B. Metabolic dysfunction-associated steatotic liver disease: a key factor in hepatocellular carcinoma therapy response. METABOLISM AND TARGET ORGAN DAMAGE 2024; 4. [DOI: 10.20517/mtod.2024.64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The conceptual evolution of non-alcoholic fatty liver disease (NAFLD) to what, since 2023, is called metabolic dysfunction-associated steatotic liver disease (MASLD) not only represents a change in the classification and definition of the disease but also reflects a broader understanding of this heterogeneous condition, which still with many aspects to refine. Although the definition of NAFLD can be interchanged to a high percentage with the new MASLD concept in different aspects, MASLD has been proposed as a relevant factor that influences the response to new immunotherapeutic treatments in the management of MASLD-related hepatocellular carcinoma (HCC), compared to HCC of other etiologies. This indicates that the etiology of HCC plays a relevant role in the prognosis, highlighting the urgency of evaluating treatment regimens for this subgroup of patients in upcoming clinical trials. A better understanding of the pathophysiology of MASLD generates strategies that not only aid in its management but also provide strategies to directly intervene in the carcinogenesis of HCC.
Collapse
|
18
|
Souza JLN, Antunes-Porto AR, da Silva Oliveira I, Amorim CCO, Pires LO, de Brito Duval I, Amaral LVBD, Souza FR, Oliveira EA, Cassali GD, Cardoso VN, Fernandes SOA, Fujiwara RT, Russo RC, Bueno LL. Screening and validating the optimal panel of housekeeping genes for 4T1 breast carcinoma and metastasis studies in mice. Sci Rep 2024; 14:26476. [PMID: 39488625 PMCID: PMC11531515 DOI: 10.1038/s41598-024-77126-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: 05/17/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
The 4T1 model is extensively employed in murine studies to elucidate the mechanisms underlying the carcinogenesis of triple-negative breast cancer. Molecular biology serves as a cornerstone in these investigations. However, accurate gene expression analyses necessitate data normalization employing housekeeping genes (HKGs) to avert spurious results. Here, we initially delve into the characteristics of the tumor evolution induced by 4T1 in mice, underscoring the imperative for additional tools for tumor monitoring and assessment methods for tracking the animals, thereby facilitating prospective studies employing this methodology. Subsequently, leveraging various software platforms, we assessed ten distinct HKGs (GAPDH, 18 S, ACTB, HPRT1, B2M, GUSB, PGK1, CCSER2, SYMPK, ANKRD17) not hitherto evaluated in the 4T1 breast cancer model, across tumors and diverse tissues afflicted by metastasis. Our principal findings underscore GAPDH as the optimal HKG for gene expression analyses in tumors, while HPRT1 emerged as the most stable in the liver and CCSER2 in the lung. These genes demonstrated consistent expression and minimal variation among experimental groups. Furthermore, employing these HKGs for normalization, we assessed TNF-α and VEGF expression in tissues and discerned significant disparities among groups. We posit that this constitutes the inaugural delineation of an ideal HKG for experiments utilizing the 4T1 model, particularly in vivo settings.
Collapse
Affiliation(s)
- Jorge Lucas Nascimento Souza
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Rafaela Antunes-Porto
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela da Silva Oliveira
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Chiara Cássia Oliveira Amorim
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Octávio Pires
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela de Brito Duval
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luisa Vitor Braga do Amaral
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Rezende Souza
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Evelyn Ane Oliveira
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Valbert Nascimento Cardoso
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Simone Odília Antunes Fernandes
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270- 901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
19
|
Seiva FRF, Agneis MLG, de Almeida MR, Caputo WL, de Souza MC, das Neves KA, Oliveira ÉN, Justulin LA, Chuffa LGDA. In Silico Analysis of Non-Conventional Oxidative Stress-Related Enzymes and Their Potential Relationship with Carcinogenesis. Antioxidants (Basel) 2024; 13:1279. [PMID: 39594421 PMCID: PMC11591236 DOI: 10.3390/antiox13111279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Carcinogenesis is driven by complex molecular events, often involving key enzymes that regulate oxidative stress (OS). While classical enzymes such as SOD, catalase, and GPx have been extensively studied, other, non-classical oxidative stress-related enzymes (OSRE) may play critical roles in cancer progression. We aimed to explore the role of OSRE involved in an OS scenario and to assess their potential contribution to carcinogenesis in some of the most prevalent cancer types. Through data mining and bioinformatic analysis of gene and protein expression and mutation data, we identified OSRE with altered expression and mutations across cancer types. Functional pathways involving EGFR, MT-ND, GST, PLCG2, PRDX6, SRC, and JAK2 were investigated. Our findings reveal that enzymes traditionally considered peripheral to OS play significant roles in tumor progression. Those OSRE may contribute to cancer initiation and progression, as well as be involved with cancer hallmarks, such as EMT and invasion, proliferation, and ROS production. In addition, enzymes like SRC and JAK2 were found to have dual roles in both promoting ROS generation and being modulated by OS. OSRE also interact with key oncogenic signaling pathways, including Wnt/β-catenin and JAK2/STAT3, linking them to cancer aggressiveness and therapeutic resistance. Future research should focus on translating these findings into clinical applications, including the development of novel inhibitors or drugs targeting these non-classical enzymes.
Collapse
Affiliation(s)
- Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Maria Luisa Gonçalves Agneis
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Matheus Ribas de Almeida
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Wesley Ladeira Caputo
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Milena Cremer de Souza
- Biological Science Center, North of Paraná State University (UENP), Bandeirantes 86360-000, PR, Brazil;
| | - Karoliny Alves das Neves
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Érika Novais Oliveira
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Luis Antônio Justulin
- Department of Structural and Functional Biology, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (L.A.J.J.); (L.G.d.A.C.)
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (L.A.J.J.); (L.G.d.A.C.)
| |
Collapse
|
20
|
Bisson WH, Liby KT, Bernard JJ. Editorial: Furthering precision medicine and cancer prevention through novel insights in molecular and chemical carcinogenesis. Front Oncol 2024; 14:1496908. [PMID: 39439960 PMCID: PMC11493655 DOI: 10.3389/fonc.2024.1496908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Affiliation(s)
- William H. Bisson
- Integrative Toxicology and Cancer Prevention, Durham, NC, United States
| | - Karen T. Liby
- Indiana University School of Medicine, Department of Medicine Division of Hematology/Oncology, Indianapolis, IN, United States
| | - Jamie J. Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Medicine, Division of Dermatology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
21
|
Faiz MB, Naeem F, Irfan M, Aslam MA, Estevinho LM, Ateşşahin DA, Alshahrani AM, Calina D, Khan K, Sharifi-Rad J. Exploring the therapeutic potential of cannabinoids in cancer by modulating signaling pathways and addressing clinical challenges. Discov Oncol 2024; 15:490. [PMID: 39331301 PMCID: PMC11436528 DOI: 10.1007/s12672-024-01356-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
For centuries, cannabinoids have been utilized for their medicinal properties, particularly in Asian and South-Asian countries. Cannabis plants, known for their psychoactive and non-psychoactive potential, were historically used for spiritual and remedial healing. However, as cannabis became predominantly a recreational drug, it faced prohibition. Recently, the therapeutic potential of cannabinoids has sparked renewed research interest, extending their use to various medical conditions, including cancer. This review aims to highlight current data on the involvement of cannabinoids in cancer signaling pathways, emphasizing their potential in cancer therapy and the need for further investigation into the underlying mechanisms. A comprehensive literature review was conducted using databases such as PubMed/MedLine, Google Scholar, Web of Science, Scopus, and Embase. The search focused on peer-reviewed articles, review articles, and clinical trials discussing the anticancer properties of cannabinoids. Inclusion criteria included studies in English on the mechanisms of action and clinical efficacy of cannabinoids in cancer. Cannabinoids, including Δ9-THC, CBD, and CBG, exhibit significant anticancer activities such as apoptosis induction, autophagy stimulation, cell cycle arrest, anti-proliferation, anti-angiogenesis, and metastasis inhibition. Clinical trials have demonstrated cannabinoids' efficacy in tumor regression and health improvement in palliative care. However, challenges such as variability in cannabinoid composition, psychoactive effects, regulatory barriers, and lack of standardized dosing remain. Cannabinoids show promising potential as anticancer agents through various mechanisms. Further large-scale, randomized controlled trials are essential to validate these findings and establish standardized therapeutic protocols. Future research should focus on elucidating detailed mechanisms, optimizing dosing, and exploring cannabinoids as primary chemotherapeutic agents.
Collapse
Affiliation(s)
- Manal Bint Faiz
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Faiza Naeem
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Muhammad Adeel Aslam
- Department of Forensic Science, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Leticia M Estevinho
- Mountain Research Center, CIMO, Polytechnic Institute of Bragança, Campus Santa Apolónia, 5300-253, Bragança, Portugal
| | - Dilek Arslan Ateşşahin
- Baskil Vocational School, Department of Plant and Animal Production, Fırat University, 23100, Elazıg, Turkey
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, College of Pharmacy, Shaqra University, Dawadimi, Saudi Arabia
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Khushbukhat Khan
- Cancer Clinical Research Unit, Trials360, Lahore, 54000, Pakistan.
| | - Javad Sharifi-Rad
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
22
|
Shenoy TN, Abdul Salam AA. Therapeutic potential of dietary bioactive compounds against anti-apoptotic Bcl-2 proteins in breast cancer. Crit Rev Food Sci Nutr 2024:1-26. [PMID: 39257284 DOI: 10.1080/10408398.2024.2398636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Breast cancer remains a leading cause of cancer-related mortality among women worldwide. One of its defining features is resistance to apoptosis, driven by aberrant expression of apoptosis-related proteins, notably the overexpression of anti-apoptotic Bcl-2 proteins. These proteins enable breast cancer cells to evade apoptosis and develop resistance to chemotherapy, underscoring their critical role as therapeutic targets. Diet plays a significant role in breast cancer risk, potentially escalating or inhibiting cancer development. Recognizing the limitations of current treatments, extensive research is focused on exploring bioactive compounds derived from natural sources such as plants, fruits, vegetables, and spices. These compounds are valued for their ability to exert potent anticancer effects with minimal toxicity and side effects. While literature extensively covers the effects of various dietary compounds in inducing apoptosis in cancer cells, comprehensive information specifically on how dietary bioactive compounds modulate anti-apoptotic Bcl-2 protein expression in breast cancer is limited. This review aims to provide a comprehensive understanding of the interaction between Bcl-2 proteins and caspases in the regulation of apoptosis, as well as the impact of dietary bioactive compounds on the modulation of anti-apoptotic Bcl-2 in breast cancer. It further explores how these interactions influence breast cancer progression and treatment outcomes.
Collapse
Affiliation(s)
- Thripthi Nagesh Shenoy
- Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Abdul Ajees Abdul Salam
- Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
23
|
Senga SS, Bisson WH, Colacci A. Key characteristics of carcinogens meet hallmarks for prevention-cutting the Gordian knot. Front Oncol 2024; 14:1420687. [PMID: 39435286 PMCID: PMC11491790 DOI: 10.3389/fonc.2024.1420687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 08/05/2024] [Indexed: 10/23/2024] Open
Abstract
The complexity of cancer requires a comprehensive approach to understand its diverse manifestations and underlying mechanisms. Initially outlined by Hanahan and Weinberg in 2000 and updated in 2010, the hallmarks of cancer provide a conceptual basis for understanding inherent variability in cancer biology. Recent expansions have further elucidated additional hallmarks, including phenotypic plasticity and senescent cells. The International Agency for Research on Cancer (IARC) has identified the key characteristics of carcinogens (KCCs) to evaluate their carcinogenic potential. We analyzed chemicals of concern for environmental exposure that interact with specific receptors to induce genomic instability, epigenetic alterations, immune suppression, and receptor-mediated effects, thereby contributing to chronic inflammation. Despite their varying degrees of carcinogenicity, these chemicals have similar KCC profiles. Our analysis highlights the pivotal role of receptor binding in activating most other KCCs, underscoring their significance in cancer initiation. Although KCCs are associated with early molecular or cellular events, they do not encompass processes directly linked to full cellular malignancy. Thus, there is a need to integrate clear endpoints that anchor KCCs to the acquisition of a complete malignant phenotype into chemical testing. From the perspective of toxicology and cancer research, an all-encompassing strategy that incorporates both existing and novel KCCs and cancer hallmarks is essential to enable the targeted identification of prevalent carcinogens and facilitate zone-specific prevention strategies. To achieve this goal, collaboration between the KCC and cancer hallmarks communities becomes essential.
Collapse
Affiliation(s)
- Sasi S. Senga
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - William H. Bisson
- Integrative Toxicology and Cancer Prevention, Durham, NC, United States
| | - Annamaria Colacci
- Agency for Prevention, Environment and Energy, Emilia-Romagna (Arpae), Bologna, Italy
- Alma Mater Institute on Healthy Planet – University of Bologna, Bologna, Italy
| |
Collapse
|
24
|
Larriba E, de Juan Romero C, García-Martínez A, Quintanar T, Rodríguez-Lescure Á, Soto JL, Saceda M, Martín-Nieto J, Barberá VM. Identification of new targets for glioblastoma therapy based on a DNA expression microarray. Comput Biol Med 2024; 179:108833. [PMID: 38981212 DOI: 10.1016/j.compbiomed.2024.108833] [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: 03/08/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
This study provides a comprehensive perspective on the deregulated pathways and impaired biological functions prevalent in human glioblastoma (GBM). In order to characterize differences in gene expression between individuals diagnosed with GBM and healthy brain tissue, we have designed and manufactured a specific, custom DNA microarray. The results obtained from differential gene expression analysis were validated by RT-qPCR. The datasets obtained from the analysis of common differential expressed genes in our cohort of patients were used to generate protein-protein interaction networks of functionally enriched genes and their biological functions. This network analysis, let us to identify 16 genes that exhibited either up-regulation (CDK4, MYC, FOXM1, FN1, E2F7, HDAC1, TNC, LAMC1, EIF4EBP1 and ITGB3) or down-regulation (PRKACB, MEF2C, CAMK2B, MAPK3, MAP2K1 and PENK) in all GBM patients. Further investigation of these genes and enriched pathways uncovered in this investigation promises to serve as a foundational step in advancing our comprehension of the molecular mechanisms underpinning GBM pathogenesis. Consequently, the present work emphasizes the critical role that the unveiled molecular pathways likely play in shaping innovative therapeutic approaches for GBM management. We finally proposed in this study a list of compounds that target hub of GBM-related genes, some of which are already in clinical use, underscoring the potential of those genes as targets for GBM treatment.
Collapse
Affiliation(s)
- Eduardo Larriba
- Human and Mammalian Genetics Group, Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, Alicante, Spain
| | - Camino de Juan Romero
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain; Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, Spain.
| | - Araceli García-Martínez
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain; Unidad de Genética Molecular, Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain
| | - Teresa Quintanar
- Servicio de Oncología Médica. Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain
| | - Álvaro Rodríguez-Lescure
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain; Servicio de Oncología Médica. Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain; School of Medicine. Universidad Miguel Hernández de Elche. Investigator, Spanish Breast Cancer Research Group (GEICAM), Spain
| | - José Luis Soto
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain; Unidad de Genética Molecular, Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain; Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, Spain
| | - José Martín-Nieto
- Human and Mammalian Genetics Group, Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, Alicante, Spain.
| | - Víctor M Barberá
- Human and Mammalian Genetics Group, Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, Alicante, Spain; Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain; Unidad de Genética Molecular, Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203, Alicante, Spain.
| |
Collapse
|
25
|
Chen Y, Chang L, Hu L, Yan C, Dai L, Shelat VG, Yarmohammadi H, Sun J. Identification of a lactylation-related gene signature to characterize subtypes of hepatocellular carcinoma using bulk sequencing data. J Gastrointest Oncol 2024; 15:1636-1646. [PMID: 39279958 PMCID: PMC11399878 DOI: 10.21037/jgo-24-405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/02/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Prior studies indicate that lactylation regulates various biological mechanisms within cancer. However, lactylation-related genes (LRGs) have been found to have limited value in predicting the prognosis of hepatocellular carcinoma (HCC). The aim of this study was to review HCC LRGs using data from The Cancer Genome Atlas (TCGA). METHODS The RNA sequencing data and related clinical information of patients with HCC patients were collected from the TCGA database. A total of 20 LRGs were selected and bioinformatics analysis was performed. A consistency cluster analysis was conducted to classify the HCC tumors. Using a lactylation-related model of HCC, prognosis, immune cell infiltration, and immunotherapy was evaluated. RESULTS A total of 4,378 genes were associated with prognosis. Twenty LRGs (i.e., ACIN1, RAN, PPP1CB, ALDOB, SUMO2, THOC2, HDAC1, SF3A1, SF3B1, HNRNPM, PPP1CC, SRRM1, PRPF6, HDAC2, H2AFV, ALYREF, H2AFZ, H2AFX, HNRNPK, and MAGOH) were identified. The 20 LRGs were used to divide TCGA-HCC patients into low-risk (G1) and high-risk (G2) categories. The upregulated genes in the G1 group primarily participate in the p53 signaling pathway, focal adhesion, extracellular matrix (ECM)-receptor interaction, and cell cycle, while the downregulated genes primarily participate in the glycolysis/gluconeogenesis, carbon metabolism, and biosynthesis of amino acids. The box plots showed a significant difference in the immune cell populations, with a higher abundance of B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, and myeloid dendritic cells in the G1 than the G2 HCC samples. Further, the box plots showed higher expression levels of seven of the eight immune checkpoint inhibitor (ICI)-related genes in the G1 HCC samples than the G2 samples. There was a significant disparity in the cancer stem cell (CSC) scores between the G1 and G2 TCGA-HCC patients. Additionally, the G1 TCGA-HCC patients had higher tumor immune dysfunction and exclusion (TIDE) scores than the G2 TCGA-HCC patients. The prognosis of the HCC patients was also predicted using a six-LRG model, comprising HDAC2, SRRM1, SF3B1, HDAC1, THOC2, and PPP1CB. CONCLUSIONS Strong correlation between LRGs and tumor classification as well as immunity in patients with HCC was identified. LRG signatures serve as reliable prognostic markers for HCC.
Collapse
Affiliation(s)
- Yan Chen
- Department of Laboratory, Yangzhou Hongquan Hospital, Yangzhou, China
| | - Li Chang
- Department of Laboratory, Yangzhou Hongquan Hospital, Yangzhou, China
| | - Ling Hu
- Department of Laboratory, Yangzhou Hongquan Hospital, Yangzhou, China
| | - Cuiping Yan
- Department of Laboratory, Yangzhou Hongquan Hospital, Yangzhou, China
| | - Liu Dai
- Department of Laboratory, Yangzhou Second People’s Hospital, Yangzhou, China
| | - Vishal G. Shelat
- Department of General Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Hooman Yarmohammadi
- Department of Interventional Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun Sun
- Department of Oncology, Yangzhou Hongquan Hospital, Yangzhou, China
| |
Collapse
|
26
|
Zhu J, Wu Y, Ji P, Qi J, Yang X, Pocha C, Tustumi F, Biachi de Castria T. Identification of a prognostic disulfidptosis-related gene signature in hepatocellular cancer. J Gastrointest Oncol 2024; 15:1647-1656. [PMID: 39279954 PMCID: PMC11399847 DOI: 10.21037/jgo-24-522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
Background Disulfidptosis regulate various biological processes in cancer. However, there is limited research on the genes related to disulfidptosis in predicting the prognosis of hepatocellular carcinoma (HCC). We aimed to develop a reliable disulfidptosis-related gene signature, which will characterize different HCC subtypes and predict their prognosis. Methods The Cancer Genome Atlas (TCGA)-HCC dataset, comprising RNA sequencing data and clinical information, was obtained from the TCGA database. The crucial disulfidptosis-related genes were selected for bioinformatic analysis in HCC. HCC tumor classification was established through a consistent cluster analysis. The prognosis and immune-cell infiltration were investigated in association with a disulfidptosis-related HCC model. Results In TCGA-HCC patients, a total of 3,621 prognostic genes and 30 key prognostic disulfidptosis-related genes were identified. Using key prognostic disulfidptosis-related genes, TCGA-HCC patients were categorized into low- and high-risk clusters. The upregulated differentially expressed genes (DEGs) in high-risk cluster 1 (C1) could significantly impact cell cycle, DNA replication, and the p53 signaling pathway, whereas the pathways associated with the downregulated DEGs in high-risk C1 could significantly impact metabolism of xenobiotics by cytochrome P450, the PPAR signaling pathway, and tyrosine metabolism. Furthermore, the immune activity of the high-risk C1 group was different to that of the low-risk cluster 2 (C2) group. The 13 disulfidptosis-related genes were finally screened using least absolute shrinkage and selection operator (LASSO) regression analysis, including ANP32E, BOP1, RPN1, SLC7A11, PPIH, PCBP2, ME1, PRDX1, FLNC, INF2, MYH11, LRPPRC, and HNRNPM. Conclusions The genes related to disulfidptosis are closely associated with tumor classification and immunity in patients with HCC. This is the first gene signature related to disulfidptosis demonstrated a strong predictive performance for the prognosis of HCC, which provide new perspectives for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Jinhong Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
- Department of Laboratory Medicine, Donggang Branch, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yan Wu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Pengyu Ji
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jun Qi
- Department of Laboratory Medicine, Donggang Branch, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xuekun Yang
- Department of Laboratory Medicine, Donggang Branch, The First Hospital of Lanzhou University, Lanzhou, China
| | - Christine Pocha
- Avera Hepatology and Transplant Institute, University of South Dakota, Sioux Falls, SD, USA
| | - Francisco Tustumi
- Department of Gastroenterology, Digestive Surgery Division, University of São Paulo, Sao Paulo, Brazil
| | - Tiago Biachi de Castria
- Moffitt Cancer Center, Tampa, FL, USA
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
27
|
Cai X, Huang W, Huang J, Zhu X, Wang L, Xia Z, Xu L. CAPZB mRNA is a novel biomarker for cervical high-grade squamous lesions. Sci Rep 2024; 14:20047. [PMID: 39209986 PMCID: PMC11362286 DOI: 10.1038/s41598-024-71112-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to evaluate the potential of capping protein (actin filament) muscle Z-line subunit β (CAPZB) messenger ribonucleic acid (mRNA) levels as a biomarker for distinguishing low-grade squamous intraepithelial lesions of the cervix (LSIL) from high-grade squamous intraepithelial lesions of the cervix (HSIL). We collected a total of 166 cervical exfoliated cells and divided them into five groups based on histopathological results. Each sample was divided into two portions, one for fluorescence in situ hybridization (FISH) detection and the other for bisulfite sequencing polymerase chain reaction (BSP) detection. We found that FISH detection of CAPZB mRNA mean fluorescence intensity (MFI) and BSP detection of CAPZB deoxyribonucleic acid (DNA) percentage of methylation rate (PMR) performed as biomarkers for distinguishing HSIL from LSIL, with an area under the receiver operating characteristic curve (AUC), sensitivity, specificity and cut-off value of 0.893, 81.25%, 80.39% and 0.616, 0.794, 64.06%, 81.37% and 0.454, respectively. Furthermore, FISH detection of CAPZB mRNA exhibited a greater AUC (0.893) for the detection of HSIL than the CAPZB DNA methylation method (0.794), indicating the CAPZB mRNA levels can be used as a biomarker for assessing cervical lesions.
Collapse
Affiliation(s)
- Xia Cai
- Department of Gynaecology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiuxiang Zhu
- Department of Gynaecology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Lifeng Wang
- Department of Gynaecology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Ziyin Xia
- Department of Gynaecology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Ling Xu
- Department of Gynaecology, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| |
Collapse
|
28
|
Shalata W, Abu Saleh O, Tourkey L, Shalata S, Neime AE, Abu Juma’a A, Soklakova A, Tourkey L, Jama AA, Yakobson A. The Efficacy of Cannabis in Oncology Patient Care and Its Anti-Tumor Effects. Cancers (Basel) 2024; 16:2909. [PMID: 39199679 PMCID: PMC11352579 DOI: 10.3390/cancers16162909] [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: 07/18/2024] [Revised: 08/11/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
As the legalization of medical cannabis expands across several countries, interest in its potential advantages among cancer patients and caregivers is burgeoning. However, patients seeking to integrate cannabis into their treatment often encounter frustration when their oncologists lack adequate information to offer guidance. This knowledge gap is exacerbated by the scarcity of published literature on the benefits of medical cannabis, leaving oncologists reliant on evidence-based data disheartened. This comprehensive narrative article, tailored for both clinicians and patients, endeavors to bridge these informational voids. It synthesizes cannabis history, pharmacology, and physiology and focuses on addressing various symptoms prevalent in cancer care, including insomnia, nausea and vomiting, appetite issues, pain management, and potential anti-cancer effects. Furthermore, by delving into the potential mechanisms of action and exploring their relevance in cancer treatment, this article aims to shed light on the potential benefits and effects of cannabis in oncology.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Omar Abu Saleh
- Department of Dermatology and Venereology, Emek Medical Centre, Afula 18341, Israel
| | - Lena Tourkey
- The Legacy Heritage Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel
| | - Ala Eddin Neime
- Medical School for International Health, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ali Abu Juma’a
- Medical School for International Health, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Arina Soklakova
- Medical School for International Health, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Lama Tourkey
- Medical School for International Health, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ashraf Abu Jama
- The Legacy Heritage Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Alexander Yakobson
- The Legacy Heritage Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
29
|
Uher O, Hadrava Vanova K, Taïeb D, Calsina B, Robledo M, Clifton-Bligh R, Pacak K. The Immune Landscape of Pheochromocytoma and Paraganglioma: Current Advances and Perspectives. Endocr Rev 2024; 45:521-552. [PMID: 38377172 PMCID: PMC11244254 DOI: 10.1210/endrev/bnae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors derived from neural crest cells from adrenal medullary chromaffin tissues and extra-adrenal paraganglia, respectively. Although the current treatment for PPGLs is surgery, optimal treatment options for advanced and metastatic cases have been limited. Hence, understanding the role of the immune system in PPGL tumorigenesis can provide essential knowledge for the development of better therapeutic and tumor management strategies, especially for those with advanced and metastatic PPGLs. The first part of this review outlines the fundamental principles of the immune system and tumor microenvironment, and their role in cancer immunoediting, particularly emphasizing PPGLs. We focus on how the unique pathophysiology of PPGLs, such as their high molecular, biochemical, and imaging heterogeneity and production of several oncometabolites, creates a tumor-specific microenvironment and immunologically "cold" tumors. Thereafter, we discuss recently published studies related to the reclustering of PPGLs based on their immune signature. The second part of this review discusses future perspectives in PPGL management, including immunodiagnostic and promising immunotherapeutic approaches for converting "cold" tumors into immunologically active or "hot" tumors known for their better immunotherapy response and patient outcomes. Special emphasis is placed on potent immune-related imaging strategies and immune signatures that could be used for the reclassification, prognostication, and management of these tumors to improve patient care and prognosis. Furthermore, we introduce currently available immunotherapies and their possible combinations with other available therapies as an emerging treatment for PPGLs that targets hostile tumor environments.
Collapse
Affiliation(s)
- Ondrej Uher
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - Katerina Hadrava Vanova
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - David Taïeb
- Department of Nuclear Medicine, CHU de La Timone, Marseille 13005, France
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Familiar Cancer Clinical Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid 28029, Spain
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, Sydney 2065, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney 2065, NSW, Australia
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| |
Collapse
|
30
|
Abbasfard Z, Behzad-Behbahani A, Rastegari B, Naeimi S, Moghanibashi M, Safari F. Overcoming Breast Cancer Drug Resistance: A Novel Approach Using siRNA-Mediated P-glycoprotein Downregulation to Enhance Vinorelbine Efficacy. Adv Pharm Bull 2024; 14:445-452. [PMID: 39206391 PMCID: PMC11347736 DOI: 10.34172/apb.2024.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/29/2024] [Accepted: 03/03/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Cancer, the second leading cause of mortality worldwide, represents a global health challenge, primarily due to drug resistance. Vinorelbine is a chemotherapeutic agent that disrupts cancer cell growth by targeting microtubules and inducing apoptosis. However, drug resistance remains a formidable obstacle. This resistance is caused by various factors including genetic mutations, drug efflux mechanisms, and DNA repair systems. Resolution of this challenge requires an innovative approach. This study investigated the potential of small interfering RNA (siRNA) to target and downregulate a vinorelbine-resistant MCF-7/ADR breast cancer cell line. Methods Cells were cultured in Dulbecco's modified Eagle's medium (DMEM) 10% fetal bovine serum/penicillin/streptomycin. An siRNA targeting ABCB1 was designed and synthesized, and the cells were transfected with siRNA at final concentrations of 10, 20, and 30 nM. The3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to assess cell viability. ABCB1 mRNA expression levels were determined by real-time polymerase chain reaction (PCR). Results MCF-7 cells exhibited a higher sensitivity to vinorelbine than MCF-7/ADR cells. MCF-7/ADR cells exhibited resistance to vinorelbine at concentrations, 12.50 and 25.00 μM. Treatment with siRNA significantly reduced ABCB1 expression by 2.93-fold (P=0.0001). Similarly, co-treatment with siRNA and vinorelbine produced a substantial 2.89-fold decrease in ABCB1 gene expression in MCF-7 cells compared to that in MCF-7/ADR cells (P=0.0001). Conclusion The results of the present study indicate that the concurrent use of siRNA and vinorelbine holds substantial promise as a therapeutic approach to overcome ABCB1-mediated multidrug resistance (MDR) in breast cancer. It is necessary to conduct comprehensive clinical trials to determine the true effectiveness of this combination therapy.
Collapse
Affiliation(s)
- Zahra Abbasfard
- Department of Genetics, College of Science, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Abbas Behzad-Behbahani
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Banafshe Rastegari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sirous Naeimi
- Department of Genetics, College of Science, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Mehdi Moghanibashi
- Department of Genetics, College of Science, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
31
|
Arizmendi-Izazaga A, Navarro-Tito N, Jiménez-Wences H, Evaristo-Priego A, Priego-Hernández VD, Dircio-Maldonado R, Zacapala-Gómez AE, Mendoza-Catalán MÁ, Illades-Aguiar B, De Nova Ocampo MA, Salmerón-Bárcenas EG, Leyva-Vázquez MA, Ortiz-Ortiz J. Bioinformatics Analysis Reveals E6 and E7 of HPV 16 Regulate Metabolic Reprogramming in Cervical Cancer, Head and Neck Cancer, and Colorectal Cancer through the PHD2-VHL-CUL2-ELOC-HIF-1α Axis. Curr Issues Mol Biol 2024; 46:6199-6222. [PMID: 38921041 PMCID: PMC11202971 DOI: 10.3390/cimb46060370] [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: 05/02/2024] [Revised: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024] Open
Abstract
Human papillomavirus 16 (HPV 16) infection is associated with several types of cancer, such as head and neck, cervical, anal, and penile cancer. Its oncogenic potential is due to the ability of the E6 and E7 oncoproteins to promote alterations associated with cell transformation. HPV 16 E6 and E7 oncoproteins increase metabolic reprogramming, one of the hallmarks of cancer, by increasing the stability of hypoxia-induced factor 1 α (HIF-1α) and consequently increasing the expression levels of their target genes. In this report, by bioinformatic analysis, we show the possible effect of HPV 16 oncoproteins E6 and E7 on metabolic reprogramming in cancer through the E6-E7-PHD2-VHL-CUL2-ELOC-HIF-1α axis. We proposed that E6 and E7 interact with VHL, CUL2, and ELOC in forming the E3 ubiquitin ligase complex that ubiquitinates HIF-1α for degradation via the proteasome. Based on the information found in the databases, it is proposed that E6 interacts with VHL by blocking its interaction with HIF-1α. On the other hand, E7 interacts with CUL2 and ELOC, preventing their binding to VHL and RBX1, respectively. Consequently, HIF-1α is stabilized and binds with HIF-1β to form the active HIF1 complex that binds to hypoxia response elements (HREs), allowing the expression of genes related to energy metabolism. In addition, we suggest an effect of E6 and E7 at the level of PHD2, VHL, CUL2, and ELOC gene expression. Here, we propose some miRNAs targeting PHD2, VHL, CUL2, and ELOC mRNAs. The effect of E6 and E7 may be the non-hydroxylation and non-ubiquitination of HIF-1α, which may regulate metabolic processes involved in metabolic reprogramming in cancer upon stabilization, non-degradation, and translocation to the nucleus.
Collapse
Affiliation(s)
- Adán Arizmendi-Izazaga
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Hilda Jiménez-Wences
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Adilene Evaristo-Priego
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Víctor Daniel Priego-Hernández
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Roberto Dircio-Maldonado
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Ana Elvira Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Miguel Ángel Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Mónica Ascención De Nova Ocampo
- Escuela Nacional de Medicina y Homeopatía, Programa Institucional de Biomedicina Molecular, Instituto Politécnico Nacional, Guillermo Massieu Helguera No. 239 Col. Fracc. La Escalera-Ticomán, Ciudad de Mexico C.P. 07320, Mexico;
| | - Eric Genaro Salmerón-Bárcenas
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México C.P. 07360, Mexico;
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (A.E.-P.); (V.D.P.-H.); (A.E.Z.-G.); (M.Á.M.-C.); (B.I.-A.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| |
Collapse
|
32
|
Bhagavatula D, Hasan TN, Vohra H, Khorami S, Hussain A. Delineating the Antiapoptotic Property of Apigenin as an Antitumor Agent: A Computational and In Vitro Study on HeLa Cells. ACS OMEGA 2024; 9:24751-24760. [PMID: 38882173 PMCID: PMC11170653 DOI: 10.1021/acsomega.4c01300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/04/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024]
Abstract
Apigenin, a flavonoid, is reported to have multiple health benefits including cancer prevention; this study evaluates the drug likeliness and Swiss ADME properties of apigenin. Apoptosis, which is a key hallmark of cancer, is associated with the deregulation of the balance between proapoptotic proteins and antiapoptotic proteins such as BCL-2,BCL-xl, BFL-1, BCL-w, BRAG-16, and MCL-1. The docking studies of apigenin with the mentioned proteins was performed to identify the interactions between the ligand and proteins, which suggested that apigenin was able to bind to most of the proteins similar to the inhibitory molecules of its native structure. A remarkable reduction in the total energy after energy minimization of apigenin-antiapoptotic protein complexes suggested increased stability of the docked complexes. The same complexes were found to be stable over a 10 ns period of molecular simulation at 300 K. These findings advocated the study to evaluate apigenin's potential to inhibit the HeLa cells at 5, 10, and 15 μM concentrations in the clonogenic assay. Apigenin inhibited the colony-forming ability of HeLa cells in a dose-dependent manner over a fortnight. Light microscopy of the treated cells displayed the morphological evidence characteristic of apoptosis in HeLa cells such as blebbing, spike formation, cytoplasmic oozing, and nuclear fragmentation. Thus, these results clearly indicate that apigenin may be used as a potential chemopreventive agent in cervical cancer management.
Collapse
Affiliation(s)
- Deepika Bhagavatula
- School of Life Sciences,Manipal Academy of Higher Education, Dubai 345050 ,United Arab Emirates
| | - Tarique Noorul Hasan
- School of Life Sciences,Manipal Academy of Higher Education, Dubai 345050 ,United Arab Emirates
- Department of Molecular Genetics, Sh. Tahnoon Bin Mohammed Medical City (STMC), Al Ain, Pure Health, Abu Dhabi 17822, United Arab Emirates
| | - Huzefa Vohra
- School of Life Sciences,Manipal Academy of Higher Education, Dubai 345050 ,United Arab Emirates
| | - Sherareh Khorami
- School of Life Sciences,Manipal Academy of Higher Education, Dubai 345050 ,United Arab Emirates
| | - Arif Hussain
- School of Life Sciences,Manipal Academy of Higher Education, Dubai 345050 ,United Arab Emirates
| |
Collapse
|
33
|
Li J, Cheng C, Zhang J. An analysis of AURKB's prognostic and immunological roles across various cancers. J Cell Mol Med 2024; 28:e18475. [PMID: 38898693 PMCID: PMC11187167 DOI: 10.1111/jcmm.18475] [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: 04/04/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Aurora kinase B (AURKB), an essential regulator in the process of mitosis, has been revealed through various studies to have a significant role in cancer development and progression. However, the specific mechanisms remain poorly understood. This study, therefore, seeks to elucidate the multifaceted role of AURKB in diverse cancer types. This study utilized bioinformatics techniques to examine the transcript, protein, promoter methylation and mutation levels of AURKB. The study further analysed associations between AURKB and factors such as prognosis, pathological stage, biological function, immune infiltration, tumour mutational burden (TMB) and microsatellite instability (MSI). In addition, immunohistochemical staining data of 50 cases of renal clear cell carcinoma and its adjacent normal tissues were collected to verify the difference in protein expression of AURKB in the two tissues. The results show that AURKB is highly expressed in most cancers, and the protein level of AURKB and the methylation level of its promoter vary among cancer types. Survival analysis showed that AURKB was associated with overall survival in 12 cancer types and progression-free survival in 11 cancer types. Elevated levels of AURKB were detected in the advanced stages of 10 different cancers. AURKB has a potential impact on cancer progression through its effects on cell cycle regulation as well as inflammatory and immune-related pathways. We observed a strong association between AURKB and immune cell infiltration, immunomodulatory factors, TMB and MSI. Importantly, we confirmed that the AURKB protein is highly expressed in kidney renal clear cell carcinoma (KIRC). Our study reveals that AURKB may be a potential biomarker for pan-cancer and KIRC.
Collapse
Affiliation(s)
- Jun Li
- Department of UrologyThe First Affiliated Hospital of Bengbu Medical UniversityBengbuChina
| | - Cui Cheng
- Department of Gynaecological OncologyThe First Affiliated Hospital of Bengbu Medical UniversityBengbuChina
| | - Jiajun Zhang
- Department of UrologyThe First Affiliated Hospital of Bengbu Medical UniversityBengbuChina
| |
Collapse
|
34
|
Yu L, Jing C, Zhuang S, Ji L, Jiang L. A novel lactylation-related gene signature for effectively distinguishing and predicting the prognosis of ovarian cancer. Transl Cancer Res 2024; 13:2497-2508. [PMID: 38881917 PMCID: PMC11170528 DOI: 10.21037/tcr-24-319] [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: 02/28/2024] [Accepted: 05/07/2024] [Indexed: 06/18/2024]
Abstract
Background Lactylation has been found to regulate several types of biological processes in cancer. However, there is limited research on lactylation-related genes in predicting the prognosis of ovarian cancer (OC). This study aimed to explore the functional roles of lactylation-related genes in OC. Methods Based on TCGA database, we obtained RNA sequencing data and clinical characteristics of patients with OC. Fourteen lactylation-related genes were screened for bioinformatic analysis in OC. Tumor classification of OC was constructed via a consistency cluster analysis. We examined the prognosis, immune-cell infiltration, and immunotherapy in relation to a lactylation-related model for OC. Results A total of 707 prognostic genes and 14 key lactylation-related genes (SNRPA1, MPHOSPH6, POLDIP3, RB1, AHNAK, MAGOHB, CALM1, EP300, HDAC1, HDAC2, HDAC3, SIRT1, SIRT2, and SIRT3) were identified in TCGA-OC patients. Based on 14 genes involved in lactylation, TCGA-OC patients were split into low-risk (G1) and high-risk (G2) groups. Downregulated differentially expressed genes (DEGs) in the low-risk G1 group were associated with thermogenesis, oxidative phosphorylation, neutrophil extracellular trap formation, and interleukin 17 (IL-17) signaling pathway, whereas upregulated DEGs were associated with proteoglycans in cancer, focal adhesion, Wnt signaling pathway, extracellular matrix (ECM)-receptor interaction, and the adherens junction. The immune activity of the low-risk G1 group was lower than that of the high-risk G2 group. Gemcitabine, bleomycin, and doxorubicin had lower half-maximal inhibitory concentration (IC50) values in the high-risk G2 patients with OC, while cisplatin and paclitaxel had higher IC50 values compared to the low-risk G1 patients. The prognosis of patients with OC was also predicted with the help of an eight-lactylation-related gene prognostic model, comprising SNRPA1, MPHOSPH6, POLDIP3, RB1, HDAC1, CALM1, HDAC2, and SIRT2. Conclusions The lactylation-related genes are closely related to tumor classification and immunity in patients with OC. There was good prognostic predictive performance for OC based on a lactylation-related signature. Our findings may offer new insights into the diagnosis and treatment of OC.
Collapse
Affiliation(s)
- Lei Yu
- Department of Obstetrics and Gynaecology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| | - Chunxia Jing
- Department of Obstetrics and Gynaecology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| | - Sulian Zhuang
- Department of Obstetrics and Gynaecology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| | - Liwei Ji
- Department of Obstetrics and Gynaecology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| | - Li Jiang
- Department of Obstetrics and Gynaecology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| |
Collapse
|
35
|
Zhuang S, Liu Z, Wu J, Yao Y, Li Z, Shen Y, Yu B, Wu D. Can O-GIcNAc Transferase (OGT) Complex Be Used as a Target for the Treatment of Hematological Malignancies? Pharmaceuticals (Basel) 2024; 17:664. [PMID: 38931332 PMCID: PMC11206344 DOI: 10.3390/ph17060664] [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: 01/27/2024] [Revised: 03/03/2024] [Accepted: 03/14/2024] [Indexed: 06/28/2024] Open
Abstract
The circulatory system is a closed conduit system throughout the body and consists of two parts as follows: the cardiovascular system and the lymphatic system. Hematological malignancies usually grow and multiply in the circulatory system, directly or indirectly affecting its function. These malignancies include multiple myeloma, leukemia, and lymphoma. O-linked β-N-acetylglucosamine (O-GlcNAc) transferase (OGT) regulates the function and stability of substrate proteins through O-GlcNAc modification. Abnormally expressed OGT is strongly associated with tumorigenesis, including hematological malignancies, colorectal cancer, liver cancer, breast cancer, and prostate cancer. In cells, OGT can assemble with a variety of proteins to form complexes to exercise related biological functions, such as OGT/HCF-1, OGT/TET, NSL, and then regulate glucose metabolism, gene transcription, cell proliferation, and other biological processes, thus affecting the development of hematological malignancies. This review summarizes the complexes involved in the assembly of OGT in cells and the role of related OGT complexes in hematological malignancies. Unraveling the complex network regulated by the OGT complex will facilitate a better understanding of hematologic malignancy development and progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Donglu Wu
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (S.Z.); (Z.L.); (J.W.); (Y.Y.); (Z.L.); (Y.S.); (B.Y.)
| |
Collapse
|
36
|
Carswell G, Chamberlin J, Bennett BD, Bushel PR, Chorley BN. Persistent gene expression and DNA methylation alterations linked to carcinogenic effects of dichloroacetic acid. Front Oncol 2024; 14:1389634. [PMID: 38764585 PMCID: PMC11099211 DOI: 10.3389/fonc.2024.1389634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/18/2024] [Indexed: 05/21/2024] Open
Abstract
Background Mechanistic understanding of transient exposures that lead to adverse health outcomes will enhance our ability to recognize biological signatures of disease. Here, we measured the transcriptomic and epigenomic alterations due to exposure to the metabolic reprogramming agent, dichloroacetic acid (DCA). Previously, we showed that exposure to DCA increased liver tumor incidence in B6C3F1 mice after continuous or early life exposures significantly over background level. Methods Using archived formalin-fixed liver samples, we utilized modern methodologies to measure gene expression and DNA methylation levels to link to previously generated phenotypic measures. Gene expression was measured by targeted RNA sequencing (TempO-seq 1500+ toxicity panel: 2754 total genes) in liver samples collected from 10-, 32-, 57-, and 78-week old mice exposed to deionized water (controls), 3.5 g/L DCA continuously in drinking water ("Direct" group), or DCA for 10-, 32-, or 57-weeks followed by deionized water until sample collection ("Stop" groups). Genome-scaled alterations in DNA methylation were measured by Reduced Representation Bisulfite Sequencing (RRBS) in 78-week liver samples for control, Direct, 10-week Stop DCA exposed mice. Results Transcriptomic changes were most robust with concurrent or adjacent timepoints after exposure was withdrawn. We observed a similar pattern with DNA methylation alterations where we noted attenuated differentially methylated regions (DMRs) in the 10-week Stop DCA exposure groups compared to the Direct group at 78-weeks. Gene pathway analysis indicated cellular effects linked to increased oxidative metabolism, a primary mechanism of action for DCA, closer to exposure windows especially early in life. Conversely, many gene signatures and pathways reversed patterns later in life and reflected more pro-tumorigenic patterns for both current and prior DCA exposures. DNA methylation patterns correlated to early gene pathway perturbations, such as cellular signaling, regulation and metabolism, suggesting persistence in the epigenome and possible regulatory effects. Conclusion Liver metabolic reprogramming effects of DCA interacted with normal age mechanisms, increasing tumor burden with both continuous and prior DCA exposure in the male B6C3F1 rodent model.
Collapse
Affiliation(s)
- Gleta Carswell
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - John Chamberlin
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Brian D. Bennett
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, United States
| | - Pierre R. Bushel
- Massive Genome Informatics Group, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, United States
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, United States
| | - Brian N. Chorley
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| |
Collapse
|
37
|
Vessella T, Xiang S, Xiao C, Stilwell M, Fok J, Shohet J, Rozen E, Zhou HS, Wen Q. DDR2 signaling and mechanosensing orchestrate neuroblastoma cell fate through different transcriptome mechanisms. FEBS Open Bio 2024; 14:867-882. [PMID: 38538106 PMCID: PMC11073507 DOI: 10.1002/2211-5463.13798] [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: 11/12/2023] [Revised: 01/24/2024] [Accepted: 03/18/2024] [Indexed: 05/07/2024] Open
Abstract
The extracellular matrix (ECM) regulates carcinogenesis by interacting with cancer cells via cell surface receptors. Discoidin Domain Receptor 2 (DDR2) is a collagen-activated receptor implicated in cell survival, growth, and differentiation. Dysregulated DDR2 expression has been identified in various cancer types, making it as a promising therapeutic target. Additionally, cancer cells exhibit mechanosensing abilities, detecting changes in ECM stiffness, which is particularly important for carcinogenesis given the observed ECM stiffening in numerous cancer types. Despite these, whether collagen-activated DDR2 signaling and ECM stiffness-induced mechanosensing exert similar effects on cancer cell behavior and whether they operate through analogous mechanisms remain elusive. To address these questions, we performed bulk RNA sequencing (RNA-seq) on human SH-SY5Y neuroblastoma cells cultured on collagen-coated substrates. Our results show that DDR2 downregulation induces significant changes in the cell transcriptome, with changes in expression of 15% of the genome, specifically affecting the genes associated with cell division and differentiation. We validated the RNA-seq results by showing that DDR2 knockdown redirects the cell fate from proliferation to senescence. Like DDR2 knockdown, increasing substrate stiffness diminishes cell proliferation. Surprisingly, RNA-seq indicates that substrate stiffness has no detectable effect on the transcriptome. Furthermore, DDR2 knockdown influences cellular responses to substrate stiffness changes, highlighting a crosstalk between these two ECM-induced signaling pathways. Based on our results, we propose that the ECM could activate DDR2 signaling and mechanosensing in cancer cells to orchestrate their cell fate through distinct mechanisms, with or without involving gene expression, thus providing novel mechanistic insights into cancer progression.
Collapse
Affiliation(s)
- Theadora Vessella
- Department of Chemical EngineeringWorcester Polytechnic InstituteMAUSA
| | | | - Cong Xiao
- Nash Family Department of Neuroscience, Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Black Family Stem Cell InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Madelyn Stilwell
- Department of Biomedical EngineeringWichita State UniversityKSUSA
| | - Jaidyn Fok
- Department of NeurobiologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Jason Shohet
- Department of PediatricsUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Esteban Rozen
- Department of PediatricsUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Crnic Institute Boulder Branch, BioFrontiers InstituteUniversity of Colorado BoulderCOUSA
| | - H. Susan Zhou
- Department of Chemical EngineeringWorcester Polytechnic InstituteMAUSA
| | - Qi Wen
- Department of PhysicsWorcester Polytechnic InstituteMAUSA
| |
Collapse
|
38
|
Sulsky SI, Greene T, Gentry PR. A framework for integrating evidence to assess hazards and risk. Crit Rev Toxicol 2024; 54:315-329. [PMID: 38808643 DOI: 10.1080/10408444.2024.2342447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/05/2024] [Indexed: 05/30/2024]
Abstract
To accurately characterize human health hazards, human, animal, and mechanistic data must be integrated and the relevance to the research question of all three lines of evidence must be considered. Mechanistic data are often critical to the full integration of animal and human data and to characterizing relevance and uncertainty. This novel evidence integration framework (EIF) provides a method for synthesizing data from comprehensive, systematic, quality-based assessments of the epidemiological and toxicological literature, including in vivo and in vitro mechanistic studies. It organizes data according to both the observed human health effects and the mechanism of action of the chemical, providing a method to support evidence synthesis. The disease-based component uses the evidence of human health outcomes studied in the best quality epidemiological literature to organize the toxicological data according to authors' stated purpose, with the pathophysiology of the disease determining the potential relevance of the toxicological data. The mechanism-based component organizes the data based on the proposed mechanisms of effect and data supporting events leading to each endpoint, with the epidemiological data potentially providing corroborating information. The EIF includes a method to cross-classify and describe the concordance of the data, and to characterize its uncertainty. At times, the two methods of organizing the data may lead to different conclusions. This facilitates identification of knowledge gaps and shows the impact of uncertainties on the strength of causal inference.
Collapse
Affiliation(s)
- Sandra I Sulsky
- Health Sciences Department, Ramboll Americas Engineering Solutions, Amherst, MA, USA
| | - Tracy Greene
- Health Sciences Department, Ramboll Americas Engineering Solutions, Monroe, LA, USA
| | - P Robinan Gentry
- Health Sciences Department, Ramboll Americas Engineering Solutions, Monroe, LA, USA
| |
Collapse
|
39
|
Carswell L, Sridharan DM, Chien LC, Hirose W, Giroux V, Nakagawa H, Pluth JM. Modeling Radiation-Induced Epithelial Cell Injury in Murine Three-Dimensional Esophageal Organoids. Biomolecules 2024; 14:519. [PMID: 38785926 PMCID: PMC11118668 DOI: 10.3390/biom14050519] [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: 01/14/2024] [Revised: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly consequence of radiation exposure to the esophagus. ESCC arises from esophageal epithelial cells that undergo malignant transformation and features a perturbed squamous cell differentiation program. Understanding the dose- and radiation quality-dependence of the esophageal epithelium response to radiation may provide insights into the ability of radiation to promote ESCC. We have explored factors that may play a role in esophageal epithelial radiosensitivity and their potential relationship to ESCC risk. We have utilized a murine three-dimensional (3D) organoid model that recapitulates the morphology and functions of the stratified squamous epithelium of the esophagus to study persistent dose- and radiation quality-dependent changes. Interestingly, although high-linear energy transfer (LET) Fe ion exposure induced a more intense and persistent alteration of squamous differentiation and 53BP1 DNA damage foci levels as compared to Cs, the MAPK/SAPK stress pathway signaling showed similar altered levels for most phospho-proteins with both radiation qualities. In addition, the lower dose of high-LET exposure also revealed nearly the same degree of morphological changes, even though only ~36% of the cells were predicted to be hit at the lower 0.1 Gy dose, suggesting that a bystander effect may be induced. Although p38 and ERK/MAPK revealed the highest levels following high-LET exposure, the findings reveal that even a low dose (0.1 Gy) of both radiation qualities can elicit a persistent stress signaling response that may critically impact the differentiation gradient of the esophageal epithelium, providing novel insights into the pathogenesis of radiation-induced esophageal injury and early stage esophageal carcinogenesis.
Collapse
Affiliation(s)
| | | | - Lung-Chang Chien
- Department of Epidemiology and Biostatistics, University of Nevada, Las Vegas, NV 89154, USA;
| | - Wataru Hirose
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; (W.H.); (H.N.)
| | - Véronique Giroux
- Department of Immunology and Cell Biology, Universite de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada;
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; (W.H.); (H.N.)
- Digestive and Liver Diseases Research Center, Organoid & Cell Culture Core, Columbia University, New York, NY 10032, USA
| | - Janice M. Pluth
- Health Physics and Diagnostic Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| |
Collapse
|
40
|
Zhou M, He X, Zhang J, Mei C, Zhong B, Ou C. tRNA-derived small RNAs in human cancers: roles, mechanisms, and clinical application. Mol Cancer 2024; 23:76. [PMID: 38622694 PMCID: PMC11020452 DOI: 10.1186/s12943-024-01992-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
Transfer RNA (tRNA)-derived small RNAs (tsRNAs) are a new type of non-coding RNAs (ncRNAs) produced by the specific cleavage of precursor or mature tRNAs. tsRNAs are involved in various basic biological processes such as epigenetic, transcriptional, post-transcriptional, and translation regulation, thereby affecting the occurrence and development of various human diseases, including cancers. Recent studies have shown that tsRNAs play an important role in tumorigenesis by regulating biological behaviors such as malignant proliferation, invasion and metastasis, angiogenesis, immune response, tumor resistance, and tumor metabolism reprogramming. These may be new potential targets for tumor treatment. Furthermore, tsRNAs can exist abundantly and stably in various bodily fluids (e.g., blood, serum, and urine) in the form of free or encapsulated extracellular vesicles, thereby affecting intercellular communication in the tumor microenvironment (TME). Meanwhile, their abnormal expression is closely related to the clinicopathological features of tumor patients, such as tumor staging, lymph node metastasis, and poor prognosis of tumor patients; thus, tsRNAs can be served as a novel type of liquid biopsy biomarker. This review summarizes the discovery, production, and expression of tsRNAs and analyzes their molecular mechanisms in tumor development and potential applications in tumor therapy, which may provide new strategies for early diagnosis and targeted therapy of tumors.
Collapse
Affiliation(s)
- Manli Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jing Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Cheng Mei
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, 410008, China.
| | - Baiyun Zhong
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
41
|
Yousuf M, Khan S, Hussain A, Alajmi MF, Shamsi A, Haque QMR, Islam A, Hassan MI. Exploring therapeutic potential of Rutin by investigating its cyclin-dependent kinase 6 inhibitory activity and binding affinity. Int J Biol Macromol 2024; 264:130624. [PMID: 38453105 DOI: 10.1016/j.ijbiomac.2024.130624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
Cyclin-dependent kinase 6 (CDK6) participates in numerous signalling pathways and regulates various physiological processes. Due to its unique structural features and promising therapeutic potential, CDK6 has emerged as a drug target for designing and developing small-molecule inhibitors for anti-cancer therapeutics and other CDK6-associated diseases. The current study evaluates binding affinity and the inhibitory potential of rutin for CDK6 to develop a proof of concept for rutin as a potent CDK6 inhibitor. Molecular docking and 200 ns all-atom simulations reveal that rutin binds to the active site pocket of CDK6, forming interactions with key residues of the binding pocket. In addition, the CDK6-rutin complex remains stable throughout the simulation trajectory. A high binding constant (Ka = 7.6 × 105M-1) indicates that rutin has a strong affinity for CDK6. Isothermal titration calorimetry has further validated a strong binding of rutin with CDK6 and its spontaneous nature. The kinase activity of CDK6 is significantly inhibited by rutin with an IC50 value of 3.10 μM. Our findings highlight the significant role of rutin in developing potential therapeutic molecules to manage cancer and CDK6-associated diseases via therapeutic targeting of CDK6.
Collapse
Affiliation(s)
- Mohd Yousuf
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shama Khan
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | | | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
42
|
Zou J, Zhang H, Wu Z, Hu W, Zhang T, Xie H, Huang Y, Zhou H. TIGD1 Is an Independent Prognostic Factor that Promotes the Progression of Colon Cancer. Cancer Biother Radiopharm 2024; 39:223-235. [PMID: 36508261 DOI: 10.1089/cbr.2022.0052] [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: 12/14/2022] Open
Abstract
Background: Trigger transposable element-derived 1 (TIGD1) is a human-specific gene, but no studies have been conducted to determine its mechanism of action. Our aim is to ascertain the function and mode of action of TIGD1 in the development of colon cancer. Materials and Methods: The authors used bioinformatics to analyze the relationship between TIGD1 and the clinical characteristics of colon cancer, as well as its prognosis. A series of cell assays were conducted to assess the function of TIGD1 in the proliferation and migration of colon cancer, and flow cytometry was used to explore its effects on apoptosis and the cell cycle. Results: The authors discovered that the expression of TIGD1 was remarkably elevated in colon cancer. Clinical correlation analysis demonstrated that TIGD1 expression was elevated in the tissues of advanced-stage patients, and it was remarkably elevated in individuals with both lymph node and distant metastasis. Further, the authors found that individuals showing elevated TIGD1 expression levels had a shortened survival time. Univariate and multivariate Cox regression analyses revealed that TIGD1 was an independent prognostic factor. Overexpression of the TIGD1 gene remarkedly enhances the proliferation and metastasis of colon cancer cells and suppresses apoptosis. In addition, the overexpression of TIGD1 can enhance the transition of tumor cells from the G1 toward the S phase. Western blot results suggested that TIGD1 may promote the malignant activity of colon cancer cells via the Wnt/β-catenin signaling pathway, Bcl-2, N-cadherin, BAX, E-cadherin, CDK6, and CyclinD1. Conclusions: TIGD1 may be an independent prognostic factor in the advancement of colon cancer, and therefore function as a therapeutic target.
Collapse
Affiliation(s)
- Junwei Zou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hesong Zhang
- Department of Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu, China
| | - Zhaoying Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Weichao Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Tingting Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hao Xie
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Yong Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hailang Zhou
- Department of Gastroenterology, Lianshui County People's Hospital, Huai'an, China
| |
Collapse
|
43
|
Chen H, Li L, Liu H, Qin P, Chen R, Liu S, Xiong H, Li Y, Yang Z, Xie M, Yang H, Jiang Q. PAX2 is regulated by estrogen/progesterone through promoter methylation in endometrioid adenocarcinoma and has an important role in carcinogenesis via the AKT/mTOR signaling pathway. J Pathol 2024; 262:467-479. [PMID: 38185904 DOI: 10.1002/path.6249] [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: 04/17/2023] [Revised: 08/08/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024]
Abstract
Endometrioid adenocarcinoma (EEC) is one of the most common cancers of the female reproductive system. In recent years, much emphasis has been placed on early diagnosis and treatment. PAX2 (Paired box 2) inactivation is reportedly an important biomarker for endometrioid intraepithelial neoplasia (EIN) and EEC. However, the role of PAX2 in EEC carcinogenesis remains unclear. PAX2 expression and associated clinical characteristics were analyzed via The Cancer Genome Atlas, Gene Expression Omnibus, and Cancer Cell Line Encyclopedia databases and clinical paired EIN/EEC tissue samples. Bioinformatic analysis was conducted to identify the putative molecular function and mechanism of PAX2. Cell proliferation, colony formation, cell migration, and invasion assays in vitro, and mouse xenograft models were utilized to study the biological functions of PAX2 in vivo. Pyrosequencing and the demethylating drug 5-Aza-dc were used to verify promoter methylation in clinical tissues and cell lines, respectively. The mechanism underlying the regulatory effect of estrogen (E2) and progesterone (P4) on PAX2 expression was investigated by receptor block assay and double luciferase reporter assay. PAX2 expression was found to be significantly downregulated in EIN and EEC tissues, its overexpression inhibited EEC cell malignant behaviors in vivo and in vitro and inhibited the AKT/mTOR signaling pathway. PAX2 inactivation in EEC was related to promoter methylation, and its expression was regulated by E2 and P4 through their receptors via promoter methylation. Our findings elucidated the expression and function of PAX2 in EEC and have provided hitherto undocumented evidence of the underlying molecular mechanisms. PAX2 expression is suppressed by estrogen prompting its methylation through estrogen receptor. Furthermore, PAX2 regulates the AKT/mTOR signaling pathway to influence EEC progression. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Lingjun Li
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
- Department of Pathology, Jingmen Central Hospital, Jingmen, PR China
| | - Huimin Liu
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Ping Qin
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Ruichao Chen
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Shaoyan Liu
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Hanzhen Xiong
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Yang Li
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Zhongfeng Yang
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Mingyu Xie
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Haili Yang
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Qingping Jiang
- Department of Pathology, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|
44
|
Kasprzak A, Geltz A. The State-of-the-Art Mechanisms and Antitumor Effects of Somatostatin in Colorectal Cancer: A Review. Biomedicines 2024; 12:578. [PMID: 38540191 PMCID: PMC10968376 DOI: 10.3390/biomedicines12030578] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/02/2024] [Accepted: 03/03/2024] [Indexed: 01/03/2025] Open
Abstract
Somatostatin, a somatotropin release inhibiting factor (SST, SRIF), is a widely distributed multifunctional cyclic peptide and acts through a transmembrane G protein-coupled receptor (SST1-SST5). Over the past decades, research has begun to reveal the molecular mechanisms underlying the anticancer activity of this hormonal peptide. Among gastrointestinal tract (GIT) tumors, direct and indirect antitumor effects of SST have been documented best in gastroenteropancreatic neuroendocrine tumors (GEP-NETs) and less well in non-endocrine cancers, including sporadic colorectal cancer (CRC). In the latter, the signaling pathways involved in the antitumor function of SST are primarily MAPK/ERK/AKT and Wnt/β-catenin. Direct (involving the MAPK pathway) and indirect (VEGF production) antiangiogenic effects of SST in CRC have also been described. The anti-inflammatory role of SST in CRC is emphasized, but detailed molecular mechanisms are still being explored. The role of SST in tumor genome/tumor microenvironment (TME)/host's gut microbiome interactions is only partially known. The results of SST analogues (SSAs)' treatment of sporadic CRC in monotherapy in vivo are not spectacular. The current review aims to present the state-of-the-art mechanisms and antitumor activity of endogenous SST and its synthetic analogues in CRC, with particular emphasis on sporadic CRC.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, University of Medical Sciences, Swiecicki Street 6, 60-781 Poznań, Poland;
| | | |
Collapse
|
45
|
Knopik-Skrocka A, Sempowicz A, Piwocka O. Plasticity and resistance of cancer stem cells as a challenge for innovative anticancer therapies - do we know enough to overcome this? EXCLI JOURNAL 2024; 23:335-355. [PMID: 38655094 PMCID: PMC11036066 DOI: 10.17179/excli2024-6972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/20/2024] [Indexed: 04/26/2024]
Abstract
According to the CSC hypothesis, cancer stem cells are pivotal in initiating, developing, and causing cancer recurrence. Since the identification of CSCs in leukemia, breast cancer, glioblastoma, and colorectal cancer in the 1990s, researchers have actively investigated the origin and biology of CSCs. However, the CSC hypothesis and the role of these cells in tumor development model is still in debate. These cells exhibit distinct surface markers, are capable of self-renewal, demonstrate unrestricted proliferation, and display metabolic adaptation. CSC phenotypic plasticity and the capacity to EMT is strictly connected to the stemness state. CSCs show high resistance to chemotherapy, radiotherapy, and immunotherapy. The plasticity of CSCs is significantly influenced by tumor microenvironment factors, such as hypoxia. Targeting the genetic and epigenetic changes of cancer cells, together with interactions with the tumor microenvironment, presents promising avenues for therapeutic strategies. See also the Graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Agnieszka Knopik-Skrocka
- Department of Cell Biology, Faculty of Biology, Adam Mickiewicz University of Poznań, Poland
- Section of Regenerative Medicine and Cancer Research, Natural Sciences Club, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznań, Poland
| | - Alicja Sempowicz
- Department of Cell Biology, Faculty of Biology, Adam Mickiewicz University of Poznań, Poland
- Section of Regenerative Medicine and Cancer Research, Natural Sciences Club, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznań, Poland
| | - Oliwia Piwocka
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Center, Poznań, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznań, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
46
|
Peng N, Liu J, Hai S, Liu Y, Zhao H, Liu W. Role of Post-Translational Modifications in Colorectal Cancer Metastasis. Cancers (Basel) 2024; 16:652. [PMID: 38339403 PMCID: PMC10854713 DOI: 10.3390/cancers16030652] [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: 01/04/2024] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract. CRC metastasis is a multi-step process with various factors involved, including genetic and epigenetic regulations, which turn out to be a serious threat to CRC patients. Post-translational modifications (PTMs) of proteins involve the addition of chemical groups, sugars, or proteins to specific residues, which fine-tunes a protein's stability, localization, or interactions to orchestrate complicated biological processes. An increasing number of recent studies suggest that dysregulation of PTMs, such as phosphorylation, ubiquitination, and glycosylation, play pivotal roles in the CRC metastasis cascade. Here, we summarized recent advances in the role of post-translational modifications in diverse aspects of CRC metastasis and its detailed molecular mechanisms. Moreover, advances in drugs targeting PTMs and their cooperation with other anti-cancer drugs, which might provide novel targets for CRC treatment and improve therapeutic efficacy, were also discussed.
Collapse
Affiliation(s)
- Na Peng
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Jingwei Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, China;
| | - Shuangshuang Hai
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Yihong Liu
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Haibo Zhao
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Weixin Liu
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| |
Collapse
|
47
|
Ijaz S, Iqbal J, Abbasi BA, Tufail A, Yaseen T, Uddin S, Usman K, Ullah R, Bibi H, Inam P, Sagindykova E, Gürer ES, Habtemariam S, Calina D, Sharifi-Rad J. Current stage of preclinical and clinical development of guggulsterone in cancers: Challenges and promises. Cell Biol Int 2024; 48:128-142. [PMID: 38148708 DOI: 10.1002/cbin.12112] [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: 09/04/2023] [Revised: 10/28/2023] [Accepted: 11/18/2023] [Indexed: 12/28/2023]
Abstract
Throughout human history, the utilization of medicinal herbs has been recognized as a crucial defense against various ailments, including cancer. Natural products with potential anticancer properties, capable of inducing apoptosis in cancer cells, have garnered substantial attention. One such agent under investigation is guggulsterone (GS), a phytosterol derived from the gum resin of the Commiphora mukul tree. This review aims to provide a comprehensive summary of recent studies elucidating the anticancer molecular mechanisms and molecular targets of GS, guiding future research and potential applications as an adjuvant drug in cancer therapy. Recent in vivo and in vitro studies have explored the biological activities of the active ingredients in Commiphora mukul. Specifically, GS emerges as a potential cancer chemopreventive and therapeutic agent. The investigations delve into the impact of GS on constitutively activated survival pathways, including Janus kinase/signal transducer and activator of transcription (JAK/STAT), nuclear factor-kappa B (NF-kB), and PI3-kinase/AKT signaling pathways. These pathways regulate antiapoptotic and proinflammatory genes, exerting control over growth and inflammatory responses. The findings highlight the potential of GS in disrupting survival pathways crucial for cancer cell viability. The inhibition of JAK/STAT, NF-kB, and PI3-kinase/AKT signaling pathways positions GS as a promising candidate for cancer therapy. The review synthesizes evidence from diverse studies, underscoring the multifaceted biological activities of GS in cancer prevention and treatment. To advance our understanding, future clinical and translational studies are imperative to determine effective doses in humans. Additionally, there is a need for the development of new pharmaceutical forms of GS to optimize therapeutic effects. This comprehensive review provides a foundation for ongoing research, offering insights into the potential of GS as a valuable addition to the armamentarium against cancer.
Collapse
Affiliation(s)
- Shumaila Ijaz
- Division of Science and Technology, Department of Botany, University of Education, Lahore, Punjab, Pakistan
| | - Javed Iqbal
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | | | - Aasma Tufail
- Division of Science and Technology, Department of Botany, University of Education, Lahore, Punjab, Pakistan
| | - Tabassum Yaseen
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Siraj Uddin
- Department of Plant Sciences, Faculty of Biological Sciences, Quaid-i-Azam University Islamabad, Islamabad, Pakistan
| | - Kiran Usman
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Rafi Ullah
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Haseena Bibi
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Palwasha Inam
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Elvira Sagindykova
- Faculty of Science and Technology, The Caspian University of Technology and Engineering Named after Sh.Yessenov, Aktau, Kazakhstan
| | - Eda Sönmez Gürer
- Department of Pharmacognosy, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Kent, UK
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | |
Collapse
|
48
|
Akbar S, Rahman A, Ahmad N, Imran M, Hafeez Z. Understanding the Role of Polyunsaturated Fatty Acids in the Development and Prevention of Cancer. Cancer Treat Res 2024; 191:57-93. [PMID: 39133404 DOI: 10.1007/978-3-031-55622-7_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] [Indexed: 08/13/2024]
Abstract
Polyunsaturated fatty acids (PUFAs), notably omega-3 (n-3) and omega-6 (n-6), have received much attention owing to their multifaceted effects not only in the management of diverse pathological conditions but also in the maintenance of overall health of an individual. A disproportionately high n-6 to n-3 ratio contributes to the development of various disorders including cancer, which ranks as a leading cause of death worldwide with profound social and economic burden. Epidemiological studies and clinical trials combined with the animal and cell culture models have demonstrated the beneficial effects of n-3 PUFAs in reducing the risk of various cancer types including breast, prostate and colon cancer. The anti-cancer actions of n-3 PUFAs are mainly attributed to their role in the modulation of a wide array of cellular processes including membrane dynamics, apoptosis, inflammation, angiogenesis, oxidative stress, gene expression and signal transduction pathways. On the contrary, n-6 PUFAs have been shown to exert pro-tumor actions; however, the inconsistent findings and controversial data emphasize upon the need to further investigation. Nevertheless, one of the biggest challenges in future is to optimize the n-6 to n-3 ratio despite the genetic predisposition, age, gender and disease severity. Moreover, a better understanding of the potential risks and benefits as well as the cellular and molecular mechanisms of the basic actions of these PUFAs is required to explore their role as adjuvants in cancer therapy. All these aspects will be reviewed in this chapter.
Collapse
Affiliation(s)
- Samina Akbar
- CALBINOTOX, Université de Lorraine, 54000, Nancy, France.
| | - Abdur Rahman
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Nazir Ahmad
- Faculty of Life Sciences, Institute of Home and Food Sciences, Government College University, Faisalabad, Pakistan
| | - Muhammad Imran
- Department of Biosciences, Faculty of Sciences, COMSATS Institute of Information Technology, Park Road, Islamabad, Pakistan
| | - Zeeshan Hafeez
- CALBINOTOX, Université de Lorraine, 54000, Nancy, France
| |
Collapse
|
49
|
Dayani L, Biganeh H, Rahimi R. Nanotechnology in the Formulation and Delivery of Natural Products for Cancer. HANDBOOK OF ONCOBIOLOGY: FROM BASIC TO CLINICAL SCIENCES 2024:1347-1369. [DOI: 10.1007/978-981-99-6263-1_68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
50
|
Malyarenko TV, Kicha AA, Kuzmich AS, Malyarenko OS, Kalinovsky AI, Popov RS, Dmitrenok PS, Ivanchina NV, Stonik VA. New Rare Triterpene Glycosides from Pacific Sun Star, Solaster pacificus, and Their Anticancer Activity. Mar Drugs 2023; 22:19. [PMID: 38248644 PMCID: PMC10820528 DOI: 10.3390/md22010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Six previously unknown triterpene glycosides, pacificusosides L-Q (1-6), and two previously known triterpene glycosides, cucumariosides B1 (7) and A5 (8), were isolated from an alcoholic extract of Pacific sun star, Solaster pacificus. The structures of 1-6 were determined using 1D and 2D NMR, ESIMS, and chemical modifications. Compound 1 is a rare type of triterpene glycoside with non-holostane aglycon, having a linear trisaccharide carbohydrate chain. Pacificusosides M-P (2-5) have new structures containing a Δ8(9)-3,16,18-trihydroxy tetracyclic triterpene moiety. This tetracyclic fragment in sea star or sea cucumber triterpene glycosides was described for the first time. All the compounds under study exhibit low or moderate cytotoxic activity against colorectal carcinoma HCT 116 cells, and breast cancer MDA-MB-231 cells were assessed by MTS assay. Compound 2 effectively suppresses the colony formation of cancer cells at a non-toxic concentration, using the soft-agar assay. A scratch assay has shown a significant anti-invasive potential of compound 2 against HCT 116 cells, but not against MDA-MB-231 cells.
Collapse
Affiliation(s)
- Timofey V. Malyarenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
- Department of Bioorganic Chemistry and Biotechnology, School of Natural Sciences, Far Eastern Federal University, Russky Island, Ajax Bay, 10, 690922 Vladivostok, Russia
| | - Alla A. Kicha
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| | - Alexandra S. Kuzmich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| | - Olesya S. Malyarenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| | - Anatoly I. Kalinovsky
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| | - Roman S. Popov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| | - Pavel S. Dmitrenok
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| | - Natalia V. Ivanchina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| | - Valentin A. Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Pr. 100-let Vladivostoku 159, 690022 Vladivostok, Russia; (A.A.K.); (A.S.K.); (O.S.M.); (A.I.K.); (R.S.P.); (P.S.D.); (N.V.I.)
| |
Collapse
|