1
|
Palabiyik AA. The role of Bcl‑2 in controlling the transition between autophagy and apoptosis (Review). Mol Med Rep 2025; 32:172. [PMID: 40242969 PMCID: PMC12045647 DOI: 10.3892/mmr.2025.13537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
The Bcl‑2 protein family serves a key role in maintaining cellular homeostasis by regulating the balance between autophagy and apoptosis. The present review aimed to summarize interactions of Bcl‑2 with key proteins, including Beclin 1, Bax and Bcl‑2 homologous antagonist/killer, as well as its influence on cellular processes such as mitophagy, nutrient sensing and endoplasmic reticulum stress response. The impact of post‑translational modifications of Bcl‑2, including phosphorylation, ubiquitination and sumoylation, is discussed with respect to their regulatory roles under stress. In pathological states, Bcl‑2 upregulation in cancer suppresses apoptosis and autophagy, thereby facilitating tumor survival and resistance to chemotherapy. Conversely, in neurodegenerative diseases, impaired autophagy and increased apoptosis contribute to neuronal loss. Therapeutic strategies targeting Bcl‑2 (for example inhibitors such as venetoclax, navitoclax, obatoclax and combination therapies involving autophagy modulators) were evaluated for their potential efficacy. There is lack of understanding of tissue‑specific functions of Bcl‑2 and its interactions with non‑coding RNAs. Future research should prioritize these areas and leverage advanced single‑cell technologies to elucidate the real‑time dynamics of Bcl‑2 in cell processes. The present review highlights the key role of Bcl‑2 in cell fate determination and highlights its potential as a therapeutic target, offering insight for the development of innovative treatments for cancer, neurodegenerative disorder and age‑related diseases.
Collapse
Affiliation(s)
- Ahmet Alperen Palabiyik
- Department of Nursing, Faculty of Health Sciences, Ardahan University, Çamlıçatak, Ardahan 75002, Turkey
| |
Collapse
|
2
|
Coussens NP, Dexheimer TS, Silvers T, Sanchez PR, Chen L, Hollingshead MG, Takebe N, Doroshow JH, Teicher BA. Combinatorial screen with apoptosis pathway targeted agents alrizomadlin, pelcitoclax, and dasminapant in multi-cell type tumor spheroids. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 33:100230. [PMID: 40210129 DOI: 10.1016/j.slasd.2025.100230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/12/2025]
Abstract
Apoptosis, or programmed cell death, plays a critical role in maintaining tissue homeostasis by eliminating damaged or abnormal cells. Dysregulation of apoptosis pathways is a hallmark of cancer, allowing malignant cells to evade cell death and proliferate uncontrollably. Targeting apoptosis pathways has emerged as a promising therapeutic strategy in cancer treatment, aiming to restore the balance between cell survival and death. The MDM2 inhibitor alrizomadlin, the Bcl-2/Bcl-xL inhibitor pelcitoclax, and the IAP family inhibitor dasminapant were evaluated both individually and in combinations with standard of care and investigational anticancer small molecules in a spheroid model of solid tumors. The multi-cell type tumor spheroids were grown from human endothelial cells and mesenchymal stem cells combined with human malignant cells that were either established or patient-derived cell lines from the NCI Patient-Derived Models Repository. The malignant cell lines were derived from a range of solid tumors including uterine carcinosarcoma, synovial sarcoma, rhabdomyosarcoma, soft tissue sarcoma, malignant fibrous histiocytoma, malignant peripheral nerve sheath tumor (MPNST), pancreas, ovary, colon, breast, and small cell lung cancer. Interactions were observed from combinations of the apoptosis pathway targeted agents. Additionally, interactions were observed from combinations of the apoptosis pathway targeted agents with other agents, including PARP inhibitors, the XPO1 inhibitor eltanexor, and the PI3K inhibitor copanlisib. Enhanced activity was also observed from combinations of the apoptosis pathway targeted agents with MAPK pathway targeted agents, including the MEK inhibitor cobimetinib as well as adagrasib and MRTX1133, which specifically target the KRAS G12C and G12D variants, respectively.
Collapse
Affiliation(s)
- Nathan P Coussens
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
| | - Thomas S Dexheimer
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Thomas Silvers
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Phillip R Sanchez
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Li Chen
- Molecular Characterization Laboratory, Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Melinda G Hollingshead
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beverly A Teicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Sarı ZB, Sarı ME, Aytar EC, Gümrükçüoğlu A, Torunoğlu EI, Ozdemir-Sanci T, Demirel G, Durmaz A. Flow Cytometry and Gene Expression Modulation by Euphorbia rigida Methanol Extract in A549 Lung Cancer Cells: Induction of Apoptosis Through Bax, Caspase-9, and Bcl-2 Pathways. Adv Biol (Weinh) 2025:e00136. [PMID: 40432491 DOI: 10.1002/adbi.202500136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/06/2025] [Indexed: 05/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) remains a major cause of cancer-related mortality. This study investigates the cytotoxic effects of Euphorbia rigida extract on A549 NSCLC cells and its potential as a therapeutic agent. Cellular morphology was observed microscopically, and cell viability was evaluated using dose-dependent proliferation assays. Apoptosis-related gene expression-including Bax, Bcl-2, and Caspase-9-was analyzed via quantitative PCR (qPCR). Chromatographic methods identified bioactive flavonoids, and molecular docking assessed their binding to cancer-related proteins. Additionally, absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiles were evaluated. The extract induced apoptotic morphological changes such as cell shrinkage and loss of intercellular contact. A dose-dependent reduction in A549 viability was observed, with an IC50 of 0.5 mg mL-1. Gene expression indicated activation of the intrinsic mitochondrial apoptotic pathway, with increased Bax and Caspase-9 and decreased Bcl-2 expression. Flow cytometry using Annexin V-allophycocyanin (V-APC) staining revealed selective cytotoxicity: significant apoptosis in A549 cells while preserving viability in BEAS-2B normal lung epithelial cells. Identified flavonoids included quercetin, apigenin, and myricetin, which showed strong binding affinities in docking studies. ADMET profiling supported their drug-likeness. These findings highlight E. rigida potential in NSCLC treatment via apoptosis induction and selective cytotoxicity.
Collapse
Affiliation(s)
- Zeynep Betül Sarı
- Faculty of Medicine, Department of Basic Medical Sciences, Medical Biology, Ankara Yıldırım Beyazıt University, Ankara, 06010, Türkiye
| | - Muhammed Emin Sarı
- Faculty of Medicine, Department of Medical Biology, Necmettin Erbakan University, Konya, 42090, Türkiye
| | - Erdi Can Aytar
- Faculty of Agriculture, Department of Horticulture, Usak University, Uşak, 64200, Türkiye
| | - Abidin Gümrükçüoğlu
- Medicinal-Aromatic Plants Application and Research Center, Artvin Çoruh University, Artvin, 08000, Türkiye
| | - Emine Incilay Torunoğlu
- Faculty of Medicine, Department of Medical Biochemistry, Necmettin Erbakan University, Konya, 42090, Türkiye
| | - Tuba Ozdemir-Sanci
- Faculty of Medicine, Department of Histology and Embriyology, Ankara Yildirim Beyazit University, Ankara, 06010, Türkiye
| | - Gamze Demirel
- Selçuk University - Akşehir Kadir Yallagöz School of Health - Department of Nutrition and Dietetics, Konya, 42560, Türkiye
| | - Alper Durmaz
- Ali Nihat Gökyiğit Botanical Garden Application and Research Center, Artvin Çoruh University, Artvin, 08000, Türkiye
| |
Collapse
|
4
|
Lemos IL, Macedo MJ, Santos FR, Montico F, Kido LA, Cagnon VHA, Maróstica Junior MR. Araticum (Annona crassiflora Mart.) by-products suppress cell proliferation and induce apoptosis particularly in androgen-dependent prostate cancer cell lines. Food Res Int 2025; 208:116124. [PMID: 40263819 DOI: 10.1016/j.foodres.2025.116124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 04/24/2025]
Abstract
Prostate cancer is the second most diagnosed type of cancer in men. The araticum (Annona crassiflora Mart.) is a fruit found in natural areas of the Brazilian cerrado, and its by-products contain a variety of compounds that have already demonstrated positive effects on cancer. To this end, we evaluated the in vitro antioxidant capacity of the extract of the peel and seed of the A. crassiflora. In addition, we investigated its antiproliferative effects and the possible mechanisms involved in inducing apoptosis in androgen-dependent and androgen-independent prostate cancer cells. The extract of A. crassiflora peel showed a high content of total phenolic compounds, reaching 222.44 mg GAE/g fdw, while the seed recorded a considerably lower value of 26.49 mg GAE/g fdw. These results indicate that the peel has a higher antioxidant capacity compared to the seed, probably due to its high content of phenolic compounds. Both extracts reduced the viability of prostate cancer cells, with the seed proving more effective. The IC50 of the seed extract was significantly lower in the PC-3 cells, presenting an IC50 of 33.24 μg/mL, 30.70 μg/mL and 24.86 μg/mL, for 24, 48 and 72 h respectively, compared to that of the peel. The peel extract showed IC50 of 277 μg/mL, 225 μg/mL and 67.30 μg/mL for the same periods. In 22Rv1 cells, the IC50 of the seed extract showed lower values, presenting IC50 of 12.64 μg/mL, 6.07 μg/mL and 5.12 μg/mL for 24, 48 and 72 h, respectively. However, the peel extract showed IC50 of 77.36 μg/mL, 42.92 μg/mL and 48.16 μg/mL for 24, 48 and 72 h. Both extracts showed a more pronounced effect on LNCaP cells. At 24 h, the IC50 of the seed extract was lower (IC50 of 22.87 μg/mL) than that of the peel extract (IC50 of 47.51 μg/mL) for LNCaP cells. However, after 48 h of treatment, the peel extract showed a decrease in IC50 of 17.64 μg/mL and the seed extract 21.13 μg/mL. However, after 72 h the seed extract was more effective in reducing cell viability with an IC50 of 6.51 μg/mL in contrast the peel showed IC50 of 11.50 μg/mL. The seed extract had a significant effect on apoptosis induction in LNCaP, increasing the protein levels of Bax, procaspase-3, caspase-9 and caspase-8, while reducing Bcl-2 and Bcl-xL expression. The seed extract also decreased the androgen receptor and PCNA levels in 22Rv1 and LNCaP cells, suggesting a possible antiproliferative mechanism mediated by the modulation of these proteins.
Collapse
Affiliation(s)
- Iara Lopes Lemos
- Department of Food Science and Nutrition (DECAN), Faculty of Food Engineering (FEA), University of Campinas (UNICAMP), Campinas, 13083-862, São Paulo, Brazil.
| | - Maria Josiane Macedo
- Department of Food Science and Nutrition (DECAN), Faculty of Food Engineering (FEA), University of Campinas (UNICAMP), Campinas, 13083-862, São Paulo, Brazil.
| | - Felipe Rabelo Santos
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP) Campinas 13083-862, São Paulo, Brazil..
| | - Fabio Montico
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP) Campinas 13083-862, São Paulo, Brazil..
| | - Larissa Akemi Kido
- Department of Food Science and Nutrition (DECAN), Faculty of Food Engineering (FEA), University of Campinas (UNICAMP), Campinas, 13083-862, São Paulo, Brazil.
| | - Valeria Helena Alves Cagnon
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP) Campinas 13083-862, São Paulo, Brazil..
| | - Mario Roberto Maróstica Junior
- Department of Food Science and Nutrition (DECAN), Faculty of Food Engineering (FEA), University of Campinas (UNICAMP), Campinas, 13083-862, São Paulo, Brazil.
| |
Collapse
|
5
|
Ruggeri RM, Grossrubatscher EM, Ciocca E, Hasballa I, Jaafar S, Oldani M, Rubino M, Russo F, Isidori AM, Colao A, Faggiano A. Incretins and SGLT-2 inhibitors in diabetic patients with neuroendocrine tumors: current updates and future directions. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09958-5. [PMID: 40175622 DOI: 10.1007/s11154-025-09958-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Neuroendocrine tumors (NET) are frequently associated with glycemic disorders, such as prediabetes or diabetes, which may result from either surgical or medical treatments or hormonal hypersecretion by the tumor itself. Moreover, pre-existing diabetes is a known risk factor for NET development, with metabolic control and antidiabetic therapies potentially influencing tumor progression. The complex interplay between diabetes and NET, which share several molecular pathways, has spurred interest in the anti-cancer effects of antidiabetic medications. This is particularly relevant as new antidiabetic drugs continue to emerge, including sodium-glucose cotransporter-2 (SGLT2) inhibitors and incretin-based therapies, such as dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide-1 receptor (GLP-1R) agonists and dual GIP/GLP- 1 R agonists. This review explores the impact of these novel pharmacological options on NET development and progression through a comprehensive analysis of pre-clinical and clinical studies, with the purpose to evaluate safety and feasibility of introducing these drugs in the treatment of NETs patients. We conducted a comprehensive search of online databases, including PubMed, ISI Web of Science, and Scopus, for studies assessing the therapeutic effects and potential mechanisms of action of incretins and SGLT2 inhibitors in patients with NET. These novel antidiabetic drugs exhibit promising anticancer properties, potentially inhibiting tumor cell proliferation and inducing apoptosis, though concerns about certain cancer risks remain. Based on current evidence, the benefits of incretin-based therapies outweigh any potential cancer risks, leading to the proposal of tailored management algorithms for diabetes in NET patients, factoring in the diabetes aetiology, comorbidities, and life expectancy.
Collapse
Affiliation(s)
- Rosaria M Ruggeri
- Endocrinology, Department of Human Pathology of Adulthood and Childhood DETEV, University of Messina, Messina, Italy
| | | | - Eleonora Ciocca
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Iderina Hasballa
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, 16132, Genoa, Italy
| | - Simona Jaafar
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Monica Oldani
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, IRCCS, Istituto Auxologico Italiano, Milan, Italy
| | - Manila Rubino
- Onco-Endocrinology Unit, European Institute of Oncology, Milan, Italy
| | - Flaminia Russo
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Sapienza University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
- UNESCO Chair "Education for Health and Sustainable Development", Federico II University, Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Sapienza University of Rome, Sant'Andrea University Hospital, Rome, Italy.
| |
Collapse
|
6
|
Li S, Ye T, Hou Z, Wang Y, Hao Z, Chen J. FOXO6: A unique transcription factor in disease regulation and therapeutic potential. Pharmacol Res 2025; 214:107691. [PMID: 40058512 DOI: 10.1016/j.phrs.2025.107691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
FOXO6, a unique member of the Forkhead box O (FOXO) transcription factor family, has emerged as a pivotal regulator in various physiological and pathological processes, including apoptosis, oxidative stress, autophagy, cell cycle control, and inflammation. Unlike other FOXO proteins, FOXO6 exhibits distinct regulatory mechanisms, particularly its inability to undergo classical nucleocytoplasmic shuttling. These unique properties suggest that FOXO6 may function through alternative pathways, positioning it as a novel research target. This review provides the first comprehensive review of FOXO6's biological functions and its roles in the progression of multiple diseases, such as cancer, metabolic disorders, neurodegenerative conditions, and cardiovascular dysfunction. We highlight FOXO6's interaction with critical signaling pathways, including PI3K/Akt, PPARγ, and TXNIP, and discuss its contributions to tumor progression, glucose and lipid metabolism, oxidative stress, and neuronal degeneration. Moreover, FOXO6's potential as a therapeutic target is explored, with particular emphasis on its ability to modulate drug resistance and its implications for disease treatment. Despite its promising therapeutic potential, the development of FOXO6-targeted therapies remains challenging due to overlapping functions within the FOXO family and the context-dependent nature of FOXO6's regulatory roles. This review underscores the need for further experimental and clinical studies to elucidate the molecular mechanisms underlying FOXO6's functions and to validate its application in disease prevention and treatment. By systematically analyzing current research, this review aims to provide a foundational reference for future studies on FOXO6, paving the way for novel therapeutic strategies targeting this unique transcription factor.
Collapse
Affiliation(s)
- Songzhe Li
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ting Ye
- The Second Hospital Affiliated Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zhitao Hou
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuqing Wang
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhihua Hao
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Chen
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
7
|
Prša P, Miller IP, Kramar B, Šuput D, Milisav I. Short-Term Fasting Induces Hepatocytes' Stress Response and Increases Their Resilience. Int J Mol Sci 2025; 26:999. [PMID: 39940770 PMCID: PMC11817670 DOI: 10.3390/ijms26030999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Fasting leads to a range of metabolic adaptations that have developed through evolution, as humans and other mammals have unequal access to food over the circadian cycle and are therefore adapted to fasting and feeding cycles. We have investigated the role of a single fasting episode in rats in triggering the stress response of liver hepatocytes. Since the stress responses were observed in both animals and isolated cells, we investigated whether the effects of the animal stressor could persist in the cells after isolation. By measuring staurosporine-induced apoptosis, stress signalling, and oxidative and antioxidant responses in hepatocytes from fasted and ad libitum-fed animals, we found that only fasting animals elicited a stress response that prevented caspase-9 activation and persisted in isolated cells. The addition of glucose oxidase, a hydrogen peroxide-producing enzyme, to the cells from ad libitum-fed animals also led to a stress response phenotype and prevented the activation of caspase-9. A single fasting episode thus leads to a stress response in normal hepatocytes, with hydrogen peroxide as a second messenger that reduces the initiation of apoptosis. This finding is the first characterisation of a mechanism underlying the effects of fasting and provides a basis for the development of methods to increase the resilience of cells. These findings need to be taken into account when interpreting the results obtained in animal and cell research models to account for the effects of overnight fasting used in many laboratory protocols. The research results also form the basis for the development of clinical applications to increase the resistance of transplants and to improve the fitness of hepatocytes under acute stress conditions in liver and some metabolic diseases.
Collapse
Affiliation(s)
- Patrik Prša
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Izak Patrik Miller
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Barbara Kramar
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Dušan Šuput
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
8
|
Akoulina EA, Bonartseva GA, Dudun AA, Kochevalina MY, Bonartsev AP, Voinova VV. Current State of Research on the Mechanisms of Biological Activity of Alginates. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S263-S286. [PMID: 40164162 DOI: 10.1134/s0006297924604519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/02/2024] [Accepted: 10/31/2024] [Indexed: 04/02/2025]
Abstract
Alginates are anionic unbranched plant and bacterial polysaccharides composed of mannuronic and guluronic acid residues. Alginates can form hydrogels under mild conditions in the presence of divalent cations (e.g., Ca2+). Because of their capacity to form gels, high biocompatibility, and relatively low cost, these polysaccharides are employed in pharmaceutical industry, medicine, food industry, cosmetology, and agriculture. Alginate oligomers produced by enzymatic cleavage of high-molecular-weight algal alginates are used as medicinal agents and dietary supplements. The global market for alginate-based products exceeds $1 billion. Alginates and their oligomers have attracted a special interest in biomedical sciences due to manifestation of various types of therapeutic activity. Across more than 50-year history of studies of alginates, over 60% scientific articles in this field have been published in the last 5 years. Unfortunately, the works dedicated to the mechanisms of biological activity of alginates and their oligosaccharides are still very scarce. This review analyzes the current state of research on the mechanisms (mainly biochemical) underlying biological and therapeutic activities of alginates (antioxidant, antibacterial, anti-inflammatory, antitumor, neuroprotective, antihypertensive, regenerative, and prebiotic). A comprehensive understanding of these mechanisms will not only improve the efficiency of alginate application in medicine and other traditional fields (cosmetology, food industry), but might also reveal their potential in new areas such as tissue engineering, nanobiotechnology, and bioelectronics.
Collapse
Affiliation(s)
- Elizaveta A Akoulina
- Biological Faculty, Shenzhen MSU-BIT University, Shenzhen, Guangdong Province, 518172, China
| | - Garina A Bonartseva
- Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Science, Moscow, 119071, Russia
| | - Andrey A Dudun
- Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Science, Moscow, 119071, Russia
- Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia
| | | | - Anton P Bonartsev
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Vera V Voinova
- Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia.
| |
Collapse
|
9
|
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
|
10
|
Zhang Y, Qin LM, Feng MF, Yu X, Wu Y. RNA-binding peptide and endosomal escape-assisting peptide (L2) improved siRNA delivery by the hexahistidine-metal assembly. J Mater Chem B 2024; 12:10309-10319. [PMID: 39282740 DOI: 10.1039/d4tb01433b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Small interfering RNAs (siRNAs), comprising 21-23 nucleotides, function by complementary binding to specific mRNA sequences, thereby suppressing target protein expression. Despite their vast potential in disease therapy, siRNAs face challenges due to their susceptibility to degradation and high electronegativity, rendering them unstable in the bloodstream and impeding their passage across endothelial barriers. Moreover, successful intracellular delivery necessitates overcoming endosomal entrapment, posing a significant hurdle for carrier material development. In this study, leveraging the strong affinity of histidine oligomers (His6) for metal ions, we engineered nanoparticles (HmA) by gentle assembly with divalent zinc ions under pH = 8 conditions. We designed the RNA-binding functional peptide L2-NTD to enhance siRNA stability and delivery efficiency when complexed with HmA. The resulting siRNA+L2-NTD@HmA nanoparticles were formed via in situ encapsulation, ensuring efficient siRNA delivery into cells with minimal cytotoxicity and degradation. This approach presents a novel strategy for the design and artificial fabrication of carriers for effective RNA delivery.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory for Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Li-Miao Qin
- State Key Laboratory for Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Meng-Fan Feng
- State Key Laboratory for Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, No. 2699 Qianjin Street, Changchun 130012, China
| | - Yuqing Wu
- State Key Laboratory for Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| |
Collapse
|
11
|
Hawash M. Advances in Cancer Therapy: A Comprehensive Review of CDK and EGFR Inhibitors. Cells 2024; 13:1656. [PMID: 39404419 PMCID: PMC11476325 DOI: 10.3390/cells13191656] [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: 08/23/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Protein kinases have essential responsibilities in controlling several cellular processes, and their abnormal regulation is strongly related to the development of cancer. The implementation of protein kinase inhibitors has significantly transformed cancer therapy by modifying treatment strategies. These inhibitors have received substantial FDA clearance in recent decades. Protein kinases have emerged as primary objectives for therapeutic interventions, particularly in the context of cancer treatment. At present, 69 therapeutics have been approved by the FDA that target approximately 24 protein kinases, which are specifically prescribed for the treatment of neoplastic illnesses. These novel agents specifically inhibit certain protein kinases, such as receptor protein-tyrosine kinases, protein-serine/threonine kinases, dual-specificity kinases, nonreceptor protein-tyrosine kinases, and receptor protein-tyrosine kinases. This review presents a comprehensive overview of novel targets of kinase inhibitors, with a specific focus on cyclin-dependent kinases (CDKs) and epidermal growth factor receptor (EGFR). The majority of the reviewed studies commenced with an assessment of cancer cell lines and concluded with a comprehensive biological evaluation of individual kinase targets. The reviewed articles provide detailed information on the structural features of potent anticancer agents and their specific activity, which refers to their ability to selectively inhibit cancer-promoting kinases including CDKs and EGFR. Additionally, the latest FDA-approved anticancer agents targeting these enzymes were highlighted accordingly.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine
| |
Collapse
|
12
|
Shi C, Li Y, You Z, Tian Y, Zhu X, Xu H, Yang M, Zhang Y, Dong R, Quan H, Shang Y, Li X. Mangiferin Ameliorates CCl 4-Triggered Acute Liver Injury by Inhibiting Inflammatory Response and Oxidative Stress: Involving the Nrf2-ARE Pathway. J Inflamm Res 2024; 17:7081-7097. [PMID: 39380664 PMCID: PMC11460351 DOI: 10.2147/jir.s476288] [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] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Purpose Acute liver injury (ALI) is characterized by inflammation and oxidative stress (OS). Although mangiferin (MGF) has antioxidant and anti-inflammatory effects, its role in ALI remains unclear. Accordingly, we investigated the MGF molecular mechanism in carbon tetrachloride (CCl4)-induced ALI in vivo and in vitro. Materials and Methods The CCl4 was utilized to induce ALI in mice. In vivo, the therapeutic effects of MGF on CCl4-induced liver injury were evaluated through biochemical assays and histomorphological analysis. Additionally, immunohistochemistry, immunofluorescence, ELISA and Western blotting were further applied to explore the mechanism. In vitro, The CCK-8 assay and flow cytometry were employed to investigate the protective effects of MGF against CCl4-induced toxicity in HepG2 cells, while mitochondrial reactive oxygen species levels and Western blotting were used to explore the biological effects and molecular mechanisms. Results MGF treatment resulted in a reduction in serum levels of AST and ALT, diminished concentrations of TNF-α, IL-6, and IL-1β in liver tissue, and concurrently decreased cellular apoptosis. Furthermore, MGF pretreatment enhanced the activity of SOD and GSH while concurrently diminishing the MDA production. This study further demonstrated the upregulation of Nrf2, NQO1, and HO-1 protein expression levels, as well as the downregulation of p-p65 protein expression levels. In vitro investigations revealed that the mitigation of CCl4-induced inflammation and OS by MGF was mediated via the Nrf2- antioxidant response element (ARE) pathway, which was disrupted by ML385 in HepG2 cells. Conclusion CCl4 can induce liver injury, while treatment with MGF mitigates ALI by inhibiting oxidative stress, inflammation, and apoptosis. The protective mechanism of MGF is mediated by the Nrf2-ARE pathway activation.
Collapse
Affiliation(s)
- Caixing Shi
- School of Basic Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yueyao Li
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Zhidong You
- School of Nursing, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yiran Tian
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Xiaoyu Zhu
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Hao Xu
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Menghan Yang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yutong Zhang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Rui Dong
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Huirong Quan
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yongyi Shang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Xiaojin Li
- School of Basic Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| |
Collapse
|
13
|
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
|
14
|
Liu Z, Yu L, Lai J, Zhang R. Decoding the molecular landscape: A novel prognostic signature for uveal melanoma unveiled through programmed cell death-associated genes. Medicine (Baltimore) 2024; 103:e38021. [PMID: 38701273 PMCID: PMC11062707 DOI: 10.1097/md.0000000000038021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
Uveal melanoma (UM) is a rare but aggressive malignant ocular tumor with a high metastatic potential and limited therapeutic options, currently lacking accurate prognostic predictors and effective individualized treatment strategies. Public databases were utilized to analyze the prognostic relevance of programmed cell death-related genes (PCDRGs) in UM transcriptomes and survival data. Consensus clustering and Lasso Cox regression analysis were performed for molecular subtyping and risk feature construction. The PCDRG-derived index (PCDI) was evaluated for its association with clinicopathological features, gene expression, drug sensitivity, and immune infiltration. A total of 369 prognostic PCDRGs were identified, which could cluster UM into 2 molecular subtypes with significant differences in prognosis and clinicopathological characteristics. Furthermore, a risk feature PCDI composed of 11 PCDRGs was constructed, capable of indicating prognosis in UM patients. Additionally, PCDI exhibited correlations with the sensitivity to 25 drugs and the infiltration of various immune cells. Enrichment analysis revealed that PCDI was associated with immune regulation-related biological processes and pathways. Finally, a nomogram for prognostic assessment of UM patients was developed based on PCDI and gender, demonstrating excellent performance. This study elucidated the potential value of PCDRGs in prognostic assessment for UM and developed a corresponding risk feature. However, further basic and clinical studies are warranted to validate the functions and mechanisms of PCDRGs in UM.
Collapse
Affiliation(s)
- Zibin Liu
- Department of Ophthalmology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lili Yu
- Department of Pediatrics, Hangzhou Linping TCM Hospital, Hangzhou, Zhejiang, China
| | - Jian Lai
- Department of Ophthalmology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Rui Zhang
- Department of Ophthalmology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|