1
|
Obisi JN, Abimbola ANJ, Babaleye OA, Atidoglo PK, Usin SG, Nwanaforo EO, Patrick-Inezi FS, Fasogbon IV, Chimezie J, Dare CA, Kuti OO, Uti DE, Omeoga HC. Unveiling the future of cancer stem cell therapy: a narrative exploration of emerging innovations. Discov Oncol 2025; 16:373. [PMID: 40120008 PMCID: PMC11929669 DOI: 10.1007/s12672-025-02102-4] [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: 11/07/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer stem cells (CSCs), are a critical subpopulation within tumours, and are defined by their capacity for self-renewal, differentiation, and tumour initiation. These unique traits contribute to tumour progression, metastasis, and resistance to conventional treatments like chemotherapy and radiotherapy, often resulting in cancer recurrence and poor patient outcomes. As such, CSCs have become focal points in developing advanced cancer therapies. This review highlights progress in CSC-targeted treatments, including chimeric antigen receptor T-cell (CAR-T) therapy, immunotherapy, molecular targeting, and nanoparticle-based drug delivery systems. Plant-derived compounds and gene-editing technologies, such as clustered regularly interspaced short palindromic repeats (CRISPR), are explored for their potential to enhance precision and minimize side effects. Metabolic pathways integral to CSC survival, such as mitochondrial dynamics, mitophagy (regulated by dynamin-related protein 1 [DRP1] and the PINK1/Parkin pathway), one-carbon metabolism, amino acid metabolism (involving enzymes like glutaminase (GLS) and glutamate dehydrogenase (GDH]), lipid metabolism, and hypoxia-induced metabolic reprogramming mediated by hypoxia-inducible factors (HIF-1α and HIF-2α), are examined as therapeutic targets. The adaptability of CSCs through autophagy, metabolic flexibility, and epigenetic regulation by metabolites like α-ketoglutarate, succinate, and fumarate is discussed. Additionally, extracellular vesicles and nicotinamide adenine dinucleotide (NAD⁺) metabolism are identified as pivotal in redox balance, DNA repair, and epigenetic modifications. Addressing challenges such as tumour heterogeneity, immune evasion, and treatment durability requires interdisciplinary collaboration. Advancing CSC-targeted therapies is essential for overcoming drug resistance and preventing cancer relapse, paving the way for transformative cancer treatments. This review underscores the importance of leveraging innovative technologies and fostering collaboration to revolutionize cancer treatment.
Collapse
Affiliation(s)
| | | | - Oluwasegun Adesina Babaleye
- Center for Human Virology and Genomics, Department of Microbiology, Nigerian Institute of Medical Research, Lagos, Nigeria
| | - Peter Kwame Atidoglo
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Saviour God'swealth Usin
- Cancer Research and Molecular Biology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Eudora Obioma Nwanaforo
- Environmental Health Science Department, School of Heath Technology, Federal University of Technology Owerri, Owerri, Nigeria
| | | | | | - Joseph Chimezie
- Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | | | - Daniel Ejim Uti
- Department of Biochemistry/Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Benue State, Nigeria.
| | | |
Collapse
|
2
|
Haque S, Mathkor DM, Bhat SA, Musayev A, Khituova L, Ramniwas S, Phillips E, Swamy N, Kumar S, Yerer MB, Tuli HS, Yadav V. A Comprehensive Review Highlighting the Prospects of Phytonutrient Berberine as an Anticancer Agent. J Biochem Mol Toxicol 2025; 39:e70073. [PMID: 39717894 DOI: 10.1002/jbt.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 10/08/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024]
Abstract
Berberine, an isoquinoline alkaloid derived from various medicinal plants, emerges as a potential therapeutic agent against diverse human diseases. It has particularly shown notable anticancer efficacy against breast, colorectal, lung, prostate, and liver cancer. Berberine results in inhibition of cancer cell proliferation, induction of apoptosis, and suppressing angiogenesis, positioning it as a versatile, multitargeted therapeutic tool against cancer. Notably, berberine enhances the effectiveness of conventional chemotherapeutic drugs, mitigating associated drug resistance. Mechanistically, it has been shown to exert its efficacy by targeting molecules like nuclear factor-kappa B (NF-κB), mitogen-activated protein kinases (MAPKs), and phosphoinositide 3-kinase (PI3K)/Akt, thereby inhibiting survival pathways and promoting apoptosis of cancer cells. Moreover, berberine influences the expression of tumor suppressor genes, curtails cancer cell migration and invasion, and modulates the tumour microenvironment. Despite promising preclinical evidence, further research is essential to comprehensively elucidate its mechanisms of action and evaluate its safety and efficacy in clinical settings. In the present review, we have highlighted the pharmacokinetics, biosynthesis, and recent research work done pertaining to berberine's strong anticancer activity. We have also emphasised on the research being done on nanoformulations of berberine, which aim to improve its stability and bioavailability.
Collapse
Affiliation(s)
- Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Sajad Ahmad Bhat
- Department of Biochemistry, International Medical School, University of International Business (UIB), Almaty, Kazakhstan
| | - Abdugani Musayev
- Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Lidiya Khituova
- Department of Pediatrics with a Course of Children's Infectious Diseases, Kazakh-Russian Medical University, Almaty, Kazakhstan
| | - Seema Ramniwas
- University Centre for Research & Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Enosh Phillips
- Department of Biotechnology, St. Aloysius' College, Jabalpur, Madhya Pradesh, India
| | - Nitin Swamy
- Department of Biotechnology, St. Aloysius' College, Jabalpur, Madhya Pradesh, India
| | - Suneel Kumar
- Department of Botany, Government Girls College, Khargone, Madhya Pradesh, India
| | - Mukerrem Betul Yerer
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
3
|
Bou Malhab LJ, Harb AA, Eldohaji L, Taneera J, Al‐Hroub HM, Abuhelwa A, Alzoubi KH, Abu‐Irmaileh B, Hudaib M, Almaliti J, Abdel‐Rahman WM, Shanableh A, Semreen MH, El‐Huneidi W, Abu‐Gharbieh E, Bustanji Y. Exploring the Anticancer Effect of Artemisia herba-alba on Colorectal Cancer: Insights From Eight Colorectal Cancer Cell Lines. Food Sci Nutr 2025; 13:e4715. [PMID: 39803277 PMCID: PMC11717013 DOI: 10.1002/fsn3.4715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent and deadly disease, necessitating the exploration of novel therapeutic strategies. Traditional chemotherapy often encounters drug resistance and adverse side effects, highlighting the need for alternative approaches. Artemisia herba-alba, a plant rich in phytochemical constituents, was investigated for its potential as an anticancer agent against colorectal cancer (CRC). The primary objective of this study was to investigate the cytotoxic effects of the methanolic extract of A. herba-alba on eight CRC cell lines including: Caco-2, DLD1, RKO+/+p53, RKO-/-p53, HCT+/+p53, HCT-/-p53, SW620, and SW480. Specifically, the study investigated the extract's impact on cell viability, apoptosis, cell cycle progression, and effects on the PI3K/AKT/mTOR signaling pathway. Chemical derivatization and Gas Chromatography-Mass Spectrometry (GC-MS) analysis revealed a diverse array of bioactive compounds, including ephedrine, hydroxyflavone, quinolinic acid, 4-hydroxybenzoic acid, borneol, β-eudesmol, and camphor, known for their cytotoxic properties. The methanolic extract of A. herba-alba exhibited varying degrees of cytotoxicity across a panel of CRC cell lines, with IC50 values indicating differential sensitivity. The extract triggered apoptosis in many cell lines, irrespective of p53 status. Importantly, A. herba-alba extract caused G2-M phase cell cycle arrest in CRC cells, accompanied by a decrease in Cyclin B1 and CDK1 expression. Furthermore, the extract demonstrated an inhibitory effect on the PI3K/AKT/mTOR pathway, crucial in cancer progression. These findings highlight the promising anticancer potential of Artemisia herba-alba as a valuable resource for innovative CRC treatments. Further research is warranted to elucidate its specific anticancer characteristics and explore its potential incorporation into future cancer therapy approaches.
Collapse
Affiliation(s)
| | - Amani A. Harb
- Department of Basic Sciences, Faculty of Arts and SciencesAl‐Ahliyya Amman UniversityAmmanJordan
| | - Leen Eldohaji
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
| | - Jalal Taneera
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
| | - Hamza M. Al‐Hroub
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
| | - Ahmad Abuhelwa
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of PharmacyUniversity of SharjahSharjahUAE
| | - Karem H. Alzoubi
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of PharmacyUniversity of SharjahSharjahUAE
| | | | | | - Jehad Almaliti
- School of PharmacyThe University of JordanAmmanJordan
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Wael M. Abdel‐Rahman
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- Department of Medical Laboratory Sciences, College of Health SciencesUniversity of SharjahSharjahUAE
| | - Abdallah Shanableh
- Research Institute of Science and Engineering (RISE)University of SharjahSharjahUAE
| | - Mohammad H. Semreen
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of PharmacyUniversity of SharjahSharjahUAE
| | - Waseem El‐Huneidi
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
| | - Eman Abu‐Gharbieh
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
- School of PharmacyThe University of JordanAmmanJordan
| | - Yasser Bustanji
- Sharjah Institute for Medical ResearchUniversity of SharjahSharjahUAE
- College of MedicineUniversity of SharjahSharjahUAE
- School of PharmacyThe University of JordanAmmanJordan
| |
Collapse
|
4
|
Demir K, Turgut R, Şentürk S, Işıklar H, Günalan E. The Therapeutic Effects of Bioactive Compounds on Colorectal Cancer via PI3K/Akt/mTOR Signaling Pathway: A Critical Review. Food Sci Nutr 2024; 12:9951-9973. [PMID: 39723045 PMCID: PMC11666977 DOI: 10.1002/fsn3.4534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 12/28/2024] Open
Abstract
Understanding the molecular signaling pathways of colorectal cancer (CRC) can be accepted as the first step in treatment strategy. Permanent mTOR signaling activation stimulates the CRC process via various biological processes. It supplies the survival of CRC stem cells, tumorigenesis, morbidity, and decreased response to drugs in CRC pathogenesis. Therefore, inhibition of the mTOR signaling by numerous bioactive components may be effective against CRC. The study aims to discuss the therapeutic capacity of various polyphenols, terpenoids, and alkaloids on CRC via the PI3K/Akt/mTOR pathway. The potential molecular effects of bioactive compounds on the mTOR pathway's upstream and downstream targets are examined. Each bioactive component causes various physiological processes, such as triggering free radical production, disruption of mitochondrial membrane potential, cell cycle arrest, inhibition of CRC stem cell migration, and suppression of glycolysis through mTOR signaling inhibition. As a result, carcinogenesis is inhibited by inducing apoptosis and autophagy. However, it should be noted that studies are primarily in vitro dose-dependent treatment researchers. This study raises awareness about the role of phenolic compounds in treating CRC, contributing to their future use as anticancer agents. These bioactive compounds have the potential to be developed into food supplementation to prevent and treat various cancer types including CRC. This review has the potential to lead to further development of clinical studies. In the future, mTOR inhibition by applying several bioactive agents using advanced drug delivery systems may contribute to CRC treatment with 3D cell culture and in vivo clinical studies.
Collapse
Affiliation(s)
- Kübra Demir
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
- Faculty of Health Science, Department of Nutrition and DieteticsSabahattin Zaim UniversityIstanbulTürkiye
| | - Rana Turgut
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Selcen Şentürk
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Handan Işıklar
- Faculty of Medicine, Department of Internal MedicineYalova UniversityYalovaTürkiye
| | - Elif Günalan
- Faculty of Health Science, Department of Nutrition and DieteticsIstanbul Health and Technology UniversityIstanbulTürkiye
| |
Collapse
|
5
|
Tarawneh N, Hamadneh L, Alshaer W, Al Bawab AQ, Bustanji Y, Abdalla S. Downregulation of aquaporins and PI3K/AKT and upregulation of PTEN expression induced by the flavone scutellarein in human colon cancer cell lines. Heliyon 2024; 10:e39402. [PMID: 39498081 PMCID: PMC11532241 DOI: 10.1016/j.heliyon.2024.e39402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 11/07/2024] Open
Abstract
Scutellarein has an anticancer potential, but the pathway of its action has not been elucidated. This study investigated scutellarein efficacy against human colorectal cancer (CRC) and explored the possible pathway of its action. MTT assay was employed to detect scutellarein effect on HT-29, SW-480, and HCT116 cells viability. Scutellarein impact on programmed cell death was studied by Annexin V-FITC/PI and its role on migration and invasion was detected by wound healing and transwell chambers. Aquaporin (AQP) 1, 3, and 5 expression before and after scutellarein treatment was approached by quantitative polymerase chain reaction (RT‒qPCR) and immunostaining while Western blotting was used to explore scutellarein effect on PI3K/AKT pathway. Scutellarein induced apoptosis and necrosis in CRC cells, thus inhibiting proliferation, migration, and invasion. Colon cancer cells exhibited positive staining for AQP 1, 3, and 5 which was downregulated by scutellarein. PI3K/AKT pathway mediating cell proliferation and growth was also modulated by scutellarein; phosphatase and tensin (PTEN) was upregulated, whereas PI3K, AKT, and p-AKT expressions were downregulated, and the downstream mTOR and phosphorylated mTOR were also suppressed at the protein level. Data indicated that scutellarein suppressed growth, migration as well as invasion of these CRC cells by downregulating the expression of AQP 1, 3, and 5 and upregulating PTEN where the latter inhibited the genes and the proteins involved in PI3K/AKT pathway. The data indicate that scutellarein is a promising therapeutic agent that inhibits growth, migration, and invasion of CRC cells by down-regulating the expression of AQP 1, 3, and 5 and by PTEN up-regulation, thus inhibiting PI3K/AKT. These molecular alterations represent potential prognostic biomarkers for the metastasis of colon cancer, where the down-regulation of AQPs enhances patient survival.
Collapse
Affiliation(s)
- Noor Tarawneh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University, Amman, 11733, Jordan
| | - Lama Hamadneh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University, Amman, 11733, Jordan
- Department of Basic Medical Sciences, Al-Balqa Applied University, Al-Salt, 19117, Jordan
| | | | - Abdel Qader Al Bawab
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University, Amman, 11733, Jordan
| | - Yasser Bustanji
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Shtaywy Abdalla
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
6
|
İPEK P, Baran A, Cebe DB, Ahmadian E, Eftekhari A, Baran MF. Antioxidant properties of allium turcicum Özhatay & cowley plant extract, its effects on the proliferation and migration of cancer cells. Front Pharmacol 2024; 15:1438634. [PMID: 39372216 PMCID: PMC11449766 DOI: 10.3389/fphar.2024.1438634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024] Open
Abstract
Cancer is a type of non-communicable disease that is responsible for numerous deaths worldwide. Cancer incidence and mortality rates are on the rise due to a combination of factors, such as a growing population, aging, and poor dietary habits. The Allium turcicum Özhatay & Cowley plant is an endemic plant in the area where it grows and is consumed by the public due to its various benefits. This endemic plant, which generally grows in high-altitude regions, is sold in bunches because it is costly, mixed with rock salt, crushed into powder, and consumed as a spice. The cytotoxic and growth-inhibitory effects of A. turcicum Özhatay & Cowley herb extract on human glioblastoma U373 cells, human colorectal carcinoma cell HCT-116, and healthy HUVEC cell lines were determined by the MTT method. After 24 and 48 h of application, logIC50 values in HUVEC, HCT-116, and U373 cells were defined as 3.737, 3.765; 3.513, 3.696, 4.476, and 4.104 μg/mL, respectively. We conducted a cell migration experiment to study the A. turcicum Özhatay & Cowley Extract (ATÖCE) impact on cancer cells' metastatic behavior. Our findings indicate that ATÖCE has an inhibitory effect on the migration potential of the cells used in the study. We conducted experiments using DPPH, ABTS, CUPRAC, and total phenolic content to assess the antioxidant properties of ATÖCE. The findings from the antioxidant activity experiments revealed an activity level of 0.20 ± 0.046 at IC50. Additionally, the total phenolic content was measured to be 0.26 ± 0.044 mg GAE/g.
Collapse
Affiliation(s)
- Polat İPEK
- Department of Physiology, Faculty of Veterinary Medicine, Dicle University, Diyarbakir, Türkiye
| | - Ayse Baran
- Department of Biology, Graduate Education Institute, Mardin Artuklu University, Mardin, Türkiye
| | - Deniz Barış Cebe
- Department of Chemistry, Batman University Faculty of Science, Batman, Türkiye
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry, Faculty of Science, Ege University, Izmir, Türkiye
- Department of Life Sciences, Engineered Biomaterials Research Center, Khazar University, Baku, Azerbaijan
| | - Mehmet Fırat Baran
- Department of Food Technology, Vocational School of Technical Sciences, Batman University, Batman, Türkiye
| |
Collapse
|
7
|
Rigillo G, Cappellucci G, Baini G, Vaccaro F, Miraldi E, Pani L, Tascedda F, Bruni R, Biagi M. Comprehensive Analysis of Berberis aristata DC. Bark Extracts: In Vitro and In Silico Evaluation of Bioaccessibility and Safety. Nutrients 2024; 16:2953. [PMID: 39275269 PMCID: PMC11397700 DOI: 10.3390/nu16172953] [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: 07/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Berberine (BER) is an alkaloid found, together with other protoberberinoids (PROTBERs), in several species used in medicines and food supplements. While some herbal preparations containing BER and PROTBERs, such as Berberis aristata DC. bark extracts, have shown promising potential for human health, their safety has not been fully assessed. Recently, the EFSA issued a call for data to deepen the pharmacokinetic and pharmacodynamic understanding of products containing BER and PROTBERs and to comprehensively assess their safety, especially when used in food supplements. In this context, new data were collected in this work by assessing: (i) the phytochemical profile of 16 different commercial B. aristata dry extracts, which are among the most widely used preparations containing BER and PROTBERs in Europe; (ii) the In Vitro and In Silico investigation of the pharmacokinetic properties of BER and PROTBERs; (iii) the In Vitro cytotoxicity of selected extracts in different human cell lines, including tests on hepatic cells in the presence of CYP450 substrates; (iv) the effects of the extracts on cancer cell migration; and (v) the In Vitro molecular effects of extracts in non-cancer human cells. Results showed that commercial B. aristata extracts contain BER as the main constituent, with jatrorrhizine as main secondary PROTBER. BER and jatrorrhizine were found to have a good bioaccessibility rate, but they interact with P-gp. B. aristata extracts showed limited cytotoxicity and minimal interaction with CYP450 substrates. Furthermore, tested extracts demonstrated inhibition of cancer cell migration and were devoid of any pro-tumoral effects in normal cells. Overall, our work provides a valuable overview to better elucidate important concerns regarding botanicals containing BER and PROTBERs.
Collapse
Affiliation(s)
- Giovanna Rigillo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
| | - Giorgio Cappellucci
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Giulia Baini
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Federica Vaccaro
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Elisabetta Miraldi
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Physical Sciences, Earth and Environment, University of Siena, 53100 Siena, Italy
| | - Luca Pani
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL 33136, USA
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Consorzio Interuniversitario Biotecnologie (CIB), 34148 Trieste, Italy
| | - Renato Bruni
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Marco Biagi
- Laboratory of Italian Society of Phytoterapy-SIFITLab, 53100 Siena, Italy
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| |
Collapse
|
8
|
Duda-Madej A, Viscardi S, Szewczyk W, Topola E. Natural Alkaloids in Cancer Therapy: Berberine, Sanguinarine and Chelerythrine against Colorectal and Gastric Cancer. Int J Mol Sci 2024; 25:8375. [PMID: 39125943 PMCID: PMC11313295 DOI: 10.3390/ijms25158375] [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: 07/18/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
The rising incidence of colorectal cancer (CRC) and gastric cancer (GC) worldwide, coupled with the limited effectiveness of current chemotherapeutic agents, has prioritized the search for new therapeutic options. Natural substances, which often exhibit cytostatic properties, hold significant promise in this area. This review evaluates the anticancer properties of three natural alkaloids-berberine, sanguinarine, and chelerythrine-against CRC and GC. In vivo and in vitro studies have demonstrated that these substances can reduce tumor volume and inhibit the epithelial-mesenchymal transition (EMT) of tumors. At the molecular level, these alkaloids disrupt key signaling pathways in cancer cells, including mTOR, MAPK, EGFR, PI3K/AKT, and NF-κB. Additionally, they exhibit immunomodulatory effects, leading to the induction of programmed cell death through both apoptosis and autophagy. Notably, these substances have shown synergistic effects when combined with classical cytostatic agents such as cyclophosphamide, 5-fluorouracil, cetuximab, and erlotinib. Furthermore, berberine has demonstrated the ability to restore sensitivity in individuals originally resistant to cisplatin GC. Given these findings, natural compounds emerge as a promising option in the chemotherapy of malignant gastrointestinal tumors, particularly in cases with limited treatment options. However, more research is necessary to fully understand their therapeutic potential.
Collapse
Affiliation(s)
- Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 4, 50-368 Wrocław, Poland
| | - Szymon Viscardi
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (W.S.); (E.T.)
| | - Wiktoria Szewczyk
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (W.S.); (E.T.)
| | - Ewa Topola
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (W.S.); (E.T.)
| |
Collapse
|
9
|
Wu C, Yang J, Ye C, Wu H, Shu W, Li R, Wang S, Lu Y, Chen H, Zhang Z, Yao Q. Berberine attenuates 5-fluorouracil-induced intestinal mucosal injury by modulating the gut microbiota without compromising its anti-tumor efficacy. Heliyon 2024; 10:e34528. [PMID: 39114045 PMCID: PMC11305238 DOI: 10.1016/j.heliyon.2024.e34528] [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: 04/04/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Background 5-Fluorouracil (5-Fu), a prominent chemotherapeutic agent for colorectal cancer (CRC) treatment, is often associated with gastrointestinal toxicities, particularly diarrhea. Our previous study demonstrated that berberine (BBR) ameliorates 5-Fu-induced intestinal mucosal injury by modulating the gut microbiota in rats. Nevertheless, the precise molecular mechanism underlying BBR's protective effect on intestinal mucosa remains elusive, and its impact on the anti-tumor efficacy of 5-Fu warrants further investigation. Methods The effect of BBR on 5-Fu-induced intestinal mucosal injury was investigated using a tumor-bearing murine model, employing H&E staining, 16 S rDNA sequencing, transcriptome sequencing, Western blot analysis, cell experiments and constructing a pseudo-germ-free tumor xenograft model. Result Our findings demonstrate that BBR alleviates intestinal mucosal damage, reduces the levels of inflammatory factors (IL-6, TNF-α, and IL-1β), and inhibits epithelial cell apoptosis in 5-Fu-treated mice without compromising 5-Fu's anti-tumor efficacy. Moreover, 16 S rDNA sequencing indicated that BBR significantly increases the abundance of Akkermansia and decreases the abundance of pathogenic bacteria Escherichia/Shigella at the genus level. Mechanistically, transcriptome sequencing and Western blot analysis confirmed that BBR upregulates PI3K/AKT/mTOR expression in the intestinal mucosa. However, this effect was not observed in tumor tissues. Notably, BBR did not demonstrate a direct protective effect on 5-Fu-treated CCD841 and SW480 cells. Additionally, BBR had no effect on the PI3K/AKT/mTOR pathway in the intestinal tissue of the 5-Fu-treated mouse model with a depleted gut microbiota. Conclusion This study indicates that BBR alleviates 5-Fu-induced intestinal mucosal injury by modulating the gut microbiota and regulating the PI3K/AKT/mTOR signaling pathway without compromising the anti-tumor efficacy of 5-Fu.
Collapse
Affiliation(s)
- Changhong Wu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jie Yang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chenxiao Ye
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hui Wu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wenxi Shu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Rongrong Li
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310012, China
| | - Sihan Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yi Lu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Department of Clinical Nutrition, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Haitao Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Integrated Traditional Chinese and Western Medicine Oncology Laboratory, Key Laboratory of Traditional Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, 310022, China
| | - Zewei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Qinghua Yao
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310005, China
| |
Collapse
|
10
|
Chang WL, Peng JY, Hong CL, Li PC, Lu FJ, Chen CH. Parecoxib and 5-Fluorouracil Synergistically Inhibit EMT and Subsequent Metastasis in Colorectal Cancer by Targeting PI3K/Akt/NF-κB Signaling. Biomedicines 2024; 12:1526. [PMID: 39062099 PMCID: PMC11274433 DOI: 10.3390/biomedicines12071526] [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: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer is one of the most common causes of cancer mortality worldwide, and innovative drugs for the treatment of colorectal cancer are continually being developed. 5-Fluorouracil (5-FU) is a common clinical chemotherapeutic drug. Acquired resistance to 5-FU is a clinical challenge in colorectal cancer treatment. Parecoxib is a selective COX-2-specific inhibitor that was demonstrated to inhibit metastasis in colorectal cancers in our previous study. This study aimed to investigate the synergistic antimetastatic activities of parecoxib to 5-FU in human colorectal cancer cells and determine the underlying mechanisms. Parecoxib and 5-FU synergistically suppressed metastasis in colorectal cancer cells. Treatment with the parecoxib/5-FU combination induced an increase in E-cadherin and decrease in β-catenin expression. The parecoxib/5-FU combination inhibited MMP-9 activity, and the NF-κB pathway was suppressed as well. Mechanistic analysis denoted that the parecoxib/5-FU combination hindered the essential molecules of the PI3K/Akt route to obstruct metastatic colorectal cancer. Furthermore, the parecoxib/5-FU combination could inhibit reactive oxygen species. Our work showed the antimetastatic capacity of the parecoxib/5-FU combination for treating colorectal cancers via the targeting of the PI3K/Akt/NF-κB pathway.
Collapse
Affiliation(s)
- Wan-Ling Chang
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Jyun-Yu Peng
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Chain-Lang Hong
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Pei-Ching Li
- Department of Anesthesiology, Chang Gung Memorial Hospital at Chiayi, No. 8, West Section of Jiapu Road, Puzi City 613016, Chiayi County, Taiwan; (W.-L.C.); (J.-Y.P.); (C.-L.H.); (P.-C.L.)
| | - Fung-Jou Lu
- Institute of Medicine, Chung Shan Medical University, No. 110, Section 1, Jianguo North Road, Taichung City 402306, Taiwan;
| | - Ching-Hsein Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, A25-303 Room, Life Sciences Hall, No. 300, Syuefu Road, National Chiayi University, Chiayi City 600355, Taiwan
| |
Collapse
|
11
|
Azimi Mohammadabadi M, Moazzeni A, Jafarzadeh L, Faraji F, Mansourabadi AH, Safari E. Aquaporins in colorectal cancer: exploring their role in tumorigenesis, metastasis, and drug response. Hum Cell 2024; 37:917-930. [PMID: 38806940 DOI: 10.1007/s13577-024-01078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Aquaporins (AQPs) are small, integral proteins facilitating water transport across plasma cell membranes in response to osmotic gradients. This family has 13 unique members (AQP0-12), which can also transport glycerol, urea, gases, and other salute small molecules. AQPs play a crucial role in the regulation of different cellular processes, including metabolism, migration, immunity, barrier function, and angiogenesis. These proteins are found to aberrantly overexpress in various cancers, including colorectal cancer (CRC). Growing evidence has explored AQPs as a potential diagnostic biomarker and therapeutic target in different cancers. However, there is no comprehensive review compiling the available information on the crucial role of AQPs in the context of colorectal cancer. This review highlights the significance of AQPs as the biomarker and regulator of tumor cells metabolism. In addition, the proliferation, angiogenesis, and metastasis of tumor cells related to AQPs expression as well as function are discussed. Understanding the AQPs prominent role in chemotherapy resistance is of great importance clinically.
Collapse
Affiliation(s)
- Maryam Azimi Mohammadabadi
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Ali Moazzeni
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Leila Jafarzadeh
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Mansourabadi
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada, Ottawa, Canada
- University of Ottawa, Brain and Mind Research Institute, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada, Ottawa, Canada
| | - Elahe Safari
- Breast Health & Cancer Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Dakkak BE, Taneera J, El-Huneidi W, Abu-Gharbieh E, Hamoudi R, Semreen MH, Soares NC, Abu-Rish EY, Alkawareek MY, Alkilany AM, Bustanji Y. Unlocking the Therapeutic Potential of BCL-2 Associated Protein Family: Exploring BCL-2 Inhibitors in Cancer Therapy. Biomol Ther (Seoul) 2024; 32:267-280. [PMID: 38589288 PMCID: PMC11063480 DOI: 10.4062/biomolther.2023.149] [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: 08/25/2023] [Revised: 11/05/2023] [Accepted: 12/05/2023] [Indexed: 04/10/2024] Open
Abstract
Apoptosis, programmed cell death pathway, is a vital physiological mechanism that ensures cellular homeostasis and overall cellular well-being. In the context of cancer, where evasion of apoptosis is a hallmark, the overexpression of anti-apoptotic proteins like Bcl2, Bcl-xL and Mcl-1 has been documented. Consequently, these proteins have emerged as promising targets for therapeutic interventions. The BCL-2 protein family is central to apoptosis and plays a significant importance in determining cellular fate serving as a critical determinant in this biological process. This review offers a comprehensive exploration of the BCL-2 protein family, emphasizing its dual nature. Specifically, certain members of this family promote cell survival (known as anti-apoptotic proteins), while others are involved in facilitating cell death (referred to as pro-apoptotic and BH3-only proteins). The potential of directly targeting these proteins is examined, particularly due to their involvement in conferring resistance to traditional cancer therapies. The effectiveness of such targeting strategies is also discussed, considering the tumor's propensity for anti-apoptotic pathways. Furthermore, the review highlights emerging research on combination therapies, where BCL-2 inhibitors are used synergistically with other treatments to enhance therapeutic outcomes. By understanding and manipulating the BCL-2 family and its associated pathways, we open doors to innovative and more effective cancer treatments, offering hope for resistant and aggressive cases.
Collapse
Affiliation(s)
- Bisan El Dakkak
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Jalal Taneera
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon 1649-016, Portugal
| | - Eman Y. Abu-Rish
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | | | | | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
13
|
Qannita RA, Alalami AI, Harb AA, Aleidi SM, Taneera J, Abu-Gharbieh E, El-Huneidi W, Saleh MA, Alzoubi KH, Semreen MH, Hudaib M, Bustanji Y. Targeting Hypoxia-Inducible Factor-1 (HIF-1) in Cancer: Emerging Therapeutic Strategies and Pathway Regulation. Pharmaceuticals (Basel) 2024; 17:195. [PMID: 38399410 PMCID: PMC10892333 DOI: 10.3390/ph17020195] [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: 12/17/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a key regulator for balancing oxygen in the cells. It is a transcription factor that regulates the expression of target genes involved in oxygen homeostasis in response to hypoxia. Recently, research has demonstrated the multiple roles of HIF-1 in the pathophysiology of various diseases, including cancer. It is a crucial mediator of the hypoxic response and regulator of oxygen metabolism, thus contributing to tumor development and progression. Studies showed that the expression of the HIF-1α subunit is significantly upregulated in cancer cells and promotes tumor survival by multiple mechanisms. In addition, HIF-1 has potential contributing roles in cancer progression, including cell division, survival, proliferation, angiogenesis, and metastasis. Moreover, HIF-1 has a role in regulating cellular metabolic pathways, particularly the anaerobic metabolism of glucose. Given its significant and potential roles in cancer development and progression, it has been an intriguing therapeutic target for cancer research. Several compounds targeting HIF-1-associated processes are now being used to treat different types of cancer. This review outlines emerging therapeutic strategies that target HIF-1 as well as the relevance and regulation of the HIF-1 pathways in cancer. Moreover, it addresses the employment of nanotechnology in developing these promising strategies.
Collapse
Affiliation(s)
- Reem A. Qannita
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ayah I. Alalami
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amani A. Harb
- Department of Basic Sciences, Faculty of Arts and Sciences, Al-Ahliyya Amman University, Amman 19111, Jordan;
| | - Shereen M. Aleidi
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| | - Jalal Taneera
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed A. Saleh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Karem H. Alzoubi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad Hudaib
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| |
Collapse
|
14
|
Dakilah I, Harb A, Abu-Gharbieh E, El-Huneidi W, Taneera J, Hamoudi R, Semreen MH, Bustanji Y. Potential of CDC25 phosphatases in cancer research and treatment: key to precision medicine. Front Pharmacol 2024; 15:1324001. [PMID: 38313315 PMCID: PMC10834672 DOI: 10.3389/fphar.2024.1324001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/04/2024] [Indexed: 02/06/2024] Open
Abstract
The global burden of cancer continues to rise, underscoring the urgency of developing more effective and precisely targeted therapies. This comprehensive review explores the confluence of precision medicine and CDC25 phosphatases in the context of cancer research. Precision medicine, alternatively referred to as customized medicine, aims to customize medical interventions by taking into account the genetic, genomic, and epigenetic characteristics of individual patients. The identification of particular genetic and molecular drivers driving cancer helps both diagnostic accuracy and treatment selection. Precision medicine utilizes sophisticated technology such as genome sequencing and bioinformatics to elucidate genetic differences that underlie the proliferation of cancer cells, hence facilitating the development of customized therapeutic interventions. CDC25 phosphatases, which play a crucial role in governing the progression of the cell cycle, have garnered significant attention as potential targets for cancer treatment. The dysregulation of CDC25 is a characteristic feature observed in various types of malignancies, hence classifying them as proto-oncogenes. The proteins in question, which operate as phosphatases, play a role in the activation of Cyclin-dependent kinases (CDKs), so promoting the advancement of the cell cycle. CDC25 inhibitors demonstrate potential as therapeutic drugs for cancer treatment by specifically blocking the activity of CDKs and modulating the cell cycle in malignant cells. In brief, precision medicine presents a potentially fruitful option for augmenting cancer research, diagnosis, and treatment, with an emphasis on individualized care predicated upon patients' genetic and molecular profiles. The review highlights the significance of CDC25 phosphatases in the advancement of cancer and identifies them as promising candidates for therapeutic intervention. This statement underscores the significance of doing thorough molecular profiling in order to uncover the complex molecular characteristics of cancer cells.
Collapse
Affiliation(s)
- Ibraheem Dakilah
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Amani Harb
- Department of Basic Sciences, Faculty of Arts and Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jalal Taneera
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Mohammed H Semreen
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
15
|
Zeng T, Ling C, Liang Y. Exploring active ingredients and mechanisms of Coptidis Rhizoma-ginger against colon cancer using network pharmacology and molecular docking. Technol Health Care 2024; 32:523-542. [PMID: 38759074 PMCID: PMC11191530 DOI: 10.3233/thc-248046] [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: 05/19/2024]
Abstract
BACKGROUND Colon cancer is the most prevalent and rapidly increasing malignancy globally. It has been suggested that some of the ingredients in the herb pair of Coptidis Rhizoma and ginger (Zingiber officinale), a traditional Chinese medicine, have potential anti-colon cancer properties. OBJECTIVE This study aimed to investigate the molecular mechanisms underlying the effects of the Coptidis Rhizoma-ginger herb pair in treating colon cancer, using an integrated approach combining network pharmacology and molecular docking. METHODS The ingredients of the herb pair Coptidis Rhizoma-ginger, along with their corresponding protein targets, were obtained from the Traditional Chinese Medicine System Pharmacology and Swiss Target Prediction databases. Target genes associated with colon cancer were retrieved from the GeneCards and OMIM databases. Then, the protein targets of the active ingredients in the herb pair were identified, and the disease-related overlapping targets were determined using the Venn online tool. The protein-protein interaction (PPI) network was constructed using STRING database and analyzed using Cytoscape 3.9.1 to identify key targets. Then, a compound-target-disease-pathway network map was constructed. The intersecting target genes were subjected to Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses for colon cancer treatment. Molecular docking was performed using the Molecular Operating Environment (MOE) software to predict the binding affinity between the key targets and active compounds. RESULTS Besides 1922 disease-related targets, 630 targets associated with 20 potential active compounds of the herb pair Coptidis Rhizoma-ginger were collected. Of these, 229 intersection targets were obtained. Forty key targets, including STAT3, Akt1, SRC, and HSP90AA1, were further analyzed using the ClueGO plugin in Cytoscape. These targets are involved in biological processes such as miRNA-mediated gene silencing, phosphatidylinositol 3-kinase (PI3K) signaling, and telomerase activity. KEGG enrichment analysis showed that PI3K-Akt and hypoxia-inducible factor 1 (HIF-1) signaling pathways were closely related to colon cancer prevention by the herb pair Coptidis Rhizoma-ginger. Ten genes (Akt1, TP53, STAT3, SRC, HSP90AA1, JAK2, CASP3, PTGS2, BCl2, and ESR1) were identified as key genes for validation through molecular docking simulation. CONCLUSIONS This study demonstrated that the herb pair Coptidis Rhizoma-ginger exerted preventive effects against colon cancer by targeting multiple genes, utilizing various active compounds, and modulating multiple pathways. These findings might provide the basis for further investigations into the molecular mechanisms underlying the therapeutic effects of Coptidis Rhizoma-ginger in colon cancer treatment, potentially leading to the development of novel drugs for combating this disease.
Collapse
Affiliation(s)
- Ting Zeng
- Institute of Systems Engineering and Collaborative Laboratory for Intelligent Science and Systems, Macau University of Science and Technology, Taipa, Macao, China
| | - Caijin Ling
- Faculty of Information Technology, Macau University of Science and Technology, Taipa, Macao, China
| | - Yong Liang
- Peng Cheng Laboratory, Shenzhen, Guangdong, China
| |
Collapse
|