1
|
Shirian FI, Karimi M, Alipour M, Salami S, Nourbakhsh M, Nekufar S, Safari-Alighiarloo N, Tavakoli-Yaraki M. Beta hydroxybutyrate induces lung cancer cell death, mitochondrial impairment and oxidative stress in a long term glucose-restricted condition. Mol Biol Rep 2024; 51:567. [PMID: 38656394 DOI: 10.1007/s11033-024-09501-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/01/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Metabolic plasticity gives cancer cells the ability to shift between signaling pathways to facilitate their growth and survival. This study investigates the role of glucose deprivation in the presence and absence of beta-hydroxybutyrate (BHB) in growth, death, oxidative stress and the stemness features of lung cancer cells. METHODS AND RESULTS A549 cells were exposed to various glucose conditions, both with and without beta-hydroxybutyrate (BHB), to evaluate their effects on apoptosis, mitochondrial membrane potential, reactive oxygen species (ROS) levels using flow cytometry, and the expression of CD133, CD44, SOX-9, and β-Catenin through Quantitative PCR. The activity of superoxide dismutase, glutathione peroxidase, and malondialdehyde was assessed using colorimetric assays. Treatment with therapeutic doses of BHB triggered apoptosis in A549 cells, particularly in cells adapted to glucose deprivation. The elevated ROS levels, combined with reduced levels of SOD and GPx, indicate that oxidative stress contributes to the cell arrest induced by BHB. Notably, BHB treatment under glucose-restricted conditions notably decreased CD133 expression, suggesting a potential inhibition of cell survival through the downregulation of CD133 levels. Additionally, the simultaneous decrease in mitochondrial membrane potential and increase in ROS levels indicate the potential for creating oxidative stress conditions to impede tumor cell growth in such environmental settings. CONCLUSION The induced cell death, oxidative stress and mitochondria impairment beside attenuated levels of cancer stem cell markers following BHB administration emphasize on the distinctive role of metabolic plasticity of cancer cells and propose possible therapeutic approaches to control cancer cell growth through metabolic fuels.
Collapse
Affiliation(s)
- Farzad Izak Shirian
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Milad Karimi
- Department of Immunology, School of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Alipour
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Siamak Salami
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Samira Nekufar
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Nahid Safari-Alighiarloo
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran.
| |
Collapse
|
2
|
Zhang Y, Li Y. β-hydroxybutyrate inhibits malignant phenotypes of prostate cancer cells through β-hydroxybutyrylation of indoleacetamide-N-methyltransferase. Cancer Cell Int 2024; 24:121. [PMID: 38555451 PMCID: PMC10981303 DOI: 10.1186/s12935-024-03277-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/19/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent cancers in men and is associated with high mortality and disability rates. β-hydroxybutyrate (BHB), a ketone body, has received increasing attention for its role in cancer. However, its role in PCa remains unclear. This study aimed to explore the mechanism and feasibility of BHB as a treatment alternative for PCa. METHODS Colony formation assay, flow cytometry, western blot assay, and transwell assays were performed to determine the effect of BHB on the proliferation and metastasis of PCa cells. Tumor sphere formation and aldehyde dehydrogenase assays were used to identify the impact of BHB or indoleacetamide-N-methyltransferase (INMT) on the stemness of PCa cells. N6-methyladenosine (m6A)-meRIP real-time reverse transcription polymerase chain reaction and dual luciferase assays were conducted to confirm INMT upregulation via the METTL3-m6A pathway. Co-IP assay was used to detect the epigenetic modification of INMT by BHB-mediated β-hydroxybutyrylation (kbhb) and screen enzymes that regulate INMT kbhb. Mouse xenograft experiments demonstrated the antitumor effects of BHB in vivo. RESULTS BHB can inhibit the proliferation, migration, and invasion of PCa cells by suppressing their stemness. Mechanistically, INMT, whose expression is upregulated by the METTL3-m6A pathway, was demonstrated to be an oncogenic gene that promotes the stem-like characteristics of PCa cells. BHB can suppress the malignant phenotypes of PCa by kbhb of INMT, which in turn inhibits INMT expression. CONCLUSIONS Our findings indicate a role of BHB in PCa metabolic therapy, thereby suggesting an epigenetic therapeutic strategy to target INMT in aggressive PCa. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, Henan, Henan, 450000, China.
| | - Yunlong Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, Henan, Henan, 450000, China
| |
Collapse
|
3
|
Roohy F, Siri M, Kohansal K, Ghalandari A, Rezaei R, Maleki MH, Shams M, Monsef A, Dastghaib S. Targeting apoptosis and unfolded protein response: the impact of β-hydroxybutyrate in clear cell renal cell carcinoma under glucose-deprived conditions. Mol Biol Rep 2024; 51:168. [PMID: 38252187 DOI: 10.1007/s11033-023-08977-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 11/13/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) plays a significant role in the mortality associated with kidney cancer. Targeting biological processes that inhibit cancer growth opens up new treatment possibilities. The unfolded protein response (UPR) and apoptosis have crucial roles in RCC progression. This study investigates the impact of β-hydroxybutyrate (BHB) on ccRCC cells under glucose deprivation resembling as a ketogenic diet. METHOD Caki-1 ccRCC cells were exposed to decreasing glucose concentrations alone or in combination with 10 or 25 mM BHB during 48 and 72 h. Cell viability was determined using MTT assay. The mRNA expression level of apoptosis-and UPR-related markers (Bcl-2, Bax, caspase 3, XBP1s, BIP, CHOP, ATF4, and ATF6) were assayed by qRT-PCR. RESULTS Cell viability experiments demonstrated that combining different doses of BHB with decreasing glucose levels initially improved cell viability after 48 h. Nevertheless, this trend reversed after 72 h, with higher impacts disclosed at 25 mM BHB. Apoptosis was induced in BHB-treated cells as caspase-3 and Bax were increased and Bcl-2 was downregulated. BHB supplementation reduced UPR-related gene expression (XBP1s, BIP, CHOP, ATF4, and ATF6), revealing a possible mechanism by which BHB affects cell survival. CONCLUSION This research emphasizes the dual effect of BHB, initially suppressing cell- survival under glucose deprivation but eventually triggering apoptosis and suppressing UPR signaling. These data highlight the intricate connection between metabolic reprogramming and cellular stress response in ccRCC. Further research is recommended to explore the potential of BHB as a therapeutic strategy for managing ccRCC.
Collapse
Affiliation(s)
- Fatemeh Roohy
- Department of Genetics, Islamic Azad University, Kazerun, Iran
| | - Morvarid Siri
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kiarash Kohansal
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Afsane Ghalandari
- Student Research Committee, Sari Branch, Islamic Azad University, Sari, Iran
| | - Roya Rezaei
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Mohammad Hasan Maleki
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mesbah Shams
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Udumula MP, Singh H, Rashid F, Poisson L, Tiwari N, Dimitrova I, Hijaz M, Gogoi R, Swenor M, Munkarah A, Giri S, Rattan R. Intermittent fasting induced ketogenesis inhibits mouse epithelial ovarian cancer by promoting antitumor T cell response. iScience 2023; 26:107839. [PMID: 37822507 PMCID: PMC10562806 DOI: 10.1016/j.isci.2023.107839] [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: 05/09/2023] [Revised: 06/28/2023] [Accepted: 09/02/2023] [Indexed: 10/13/2023] Open
Abstract
In various cancer models, dietary interventions have been shown to inhibit tumor growth, improve anticancer drug efficacy, and enhance immunity, but no such evidence exists for epithelial ovarian cancer (EOC), the most lethal gynecologic cancer. The anticancer immune responses induced by 16-h intermittent fasting (IF) were studied in mice with EOC. IF consistently reduced metabolic growth factors and cytokines that stimulate tumor growth, creating a tumor-hostile environment. Immune profiling showed that IF dramatically alters anti-cancer immunity by increasing CD4+ and CD8+ cells, Th1 and cytotoxic responses, and metabolic fitness. β-hydroxy butyrate (BHB), a bioactive metabolite produced by IF, partially imitates its anticancer effects by inducing CD8+ effector function. In a direct comparison, IF outperformed exogenous BHB treatment in survival and anti-tumor immune response, probably due to increased ketogenesis. Thus, IF and one of its metabolic mediators BHB suppress EOC growth and sustain a potent anti-tumor T cell response.
Collapse
Affiliation(s)
- Mary Priyanka Udumula
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Harshit Singh
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Faraz Rashid
- Metabolomics Core, Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Laila Poisson
- Department of Public Health Services and Center for Bioinformatics and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Nivedita Tiwari
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Irina Dimitrova
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Miriana Hijaz
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Radhika Gogoi
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Margaret Swenor
- Department of Lifestyle and Functional Medicine, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Adnan Munkarah
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
| | - Shailendra Giri
- Metabolomics Core, Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Ramandeep Rattan
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
- Department of Ob/Gyn, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
5
|
Udumula MP, Singh H, Faraz R, Poisson L, Tiwari N, Dimitrova I, Hijaz M, Gogoi R, Swenor M, Munkarah A, Giri S, Rattan R. Intermittent Fasting induced ketogenesis inhibits mouse epithelial ovarian tumors by promoting anti-tumor T cell response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.08.531740. [PMID: 36945428 PMCID: PMC10028914 DOI: 10.1101/2023.03.08.531740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Epithelial Ovarian Cancer (EOC) is the most lethal gynecologic cancer with limited genetic alterations identified that can be therapeutically targeted. In tumor bearing mice, short-term fasting, fasting mimicking diet and calorie restriction enhance the activity of antineoplastic treatment by modulating systemic metabolism and boosting anti-tumor immunity. We tested the outcome of sixteen-hour intermittent fasting (IF) on mouse EOC progression with focus on fasting driven antitumor immune responses. IF resulted in consistent decrease of tumor promoting metabolic growth factors and cytokines, recapitulating changes that creates a tumor antagonizing environment. Immune profiling revealed that IF profoundly reshapes anti-cancer immunity by inducing increase in CD4+ and CD8+ cells, paralleled by enhanced antitumor Th1 and cytotoxic responses, by enhancing their metabolic fitness. Metabolic studies revealed that IF generated bioactive metabolite BHB which can be a potential substitute for simulating the antitumor benefits of IF. However, in a direct comparison, IF surpassed exogenous BHB therapy in improving survival and activating anti-tumor immune response. Thus, our data provides strong evidence for IF and its metabolic mediator BHB for ameliorating EOC progression and as a viable approach in maintaining and sustaining an effective anti-tumor T cell response.
Collapse
Affiliation(s)
- Mary Priyanka Udumula
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
| | - Harshit Singh
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
| | - Rashid Faraz
- Metabolomics Core, Department of Neurology, Henry Ford Hospital, Detroit, MI 48202
| | - Laila Poisson
- Department of Public Health Services and Center for Bioinformatics and Henry Ford Cancer Institute, Detroit, MI
| | - Nivedita Tiwari
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
| | - Irina Dimitrova
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
| | - Miriana Hijaz
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
| | - Radhika Gogoi
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI
| | - Margaret Swenor
- Department of Lifestyle and Functional Medicine, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
| | - Adnan Munkarah
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
| | - Shailendra Giri
- Metabolomics Core, Department of Neurology, Henry Ford Hospital, Detroit, MI 48202
| | - Ramandeep Rattan
- Department of Women’s Health Services, Henry Ford Hospital and Henry Ford Cancer Institute, Detroit, MI
- Department of Oncology, Wayne State University, Detroit, MI
| |
Collapse
|
6
|
Ulm MA, Redfern TM, Wilson BR, Ponnusamy S, Asemota S, Blackburn PW, Wang Y, ElNaggar AC, Narayanan R. Integrin-Linked Kinase Is a Novel Therapeutic Target in Ovarian Cancer. J Pers Med 2020; 10:jpm10040246. [PMID: 33256002 PMCID: PMC7712057 DOI: 10.3390/jpm10040246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE The objective of this study is to identify and validate novel therapeutic target(s) in ovarian cancer. BACKGROUND Development of targeted therapeutics in ovarian cancer has been limited by molecular heterogeneity. Although gene expression datasets are available, most of them lack appropriate pair-matched controls to define the alterations that result in the transformation of normal ovarian cells to cancerous cells. METHODS We used microarray to compare the gene expression of treatment-naïve ovarian cancer tissue samples to pair-matched normal adjacent ovarian tissue from 24 patients. Ingenuity Pathway Analysis (IPA) was used to identify target pathways for further analysis. Integrin-linked kinase (ILK) expression in SKOV3 and OV90 cells was determined using Western blot. ILK was knocked down using CRISPR/Cas9 constructs. Subcutaneous xenograft study to determine the effect of ILK knockdown on tumor growth was performed in NOD SCID gamma mice. RESULTS Significant upregulation of the ILK pathway was identified in 22 of the 24 cancer specimens, identifying it as a potential player that could contribute to the transformation of normal ovarian cells to cancerous cells. Knockdown of ILK in SKOV3 cells resulted in decreased cell proliferation and tumor growth, and inhibition of downstream kinase, AKT (protein kinase B). These results were further validated using an ILK-1 chemical inhibitor, compound 22. CONCLUSION Our initial findings validate ILK as a potential therapeutic target for molecular inhibition in ovarian cancer, which warrants further investigation.
Collapse
Affiliation(s)
- Michael A. Ulm
- Division of Gynecologic Oncology, West Cancer Center and Research Institute, Memphis, TN 38138, USA; (M.A.U.); (T.M.R.); (B.R.W.); (P.W.B.); (A.C.E.)
| | - Tiffany M. Redfern
- Division of Gynecologic Oncology, West Cancer Center and Research Institute, Memphis, TN 38138, USA; (M.A.U.); (T.M.R.); (B.R.W.); (P.W.B.); (A.C.E.)
| | - Ben R. Wilson
- Division of Gynecologic Oncology, West Cancer Center and Research Institute, Memphis, TN 38138, USA; (M.A.U.); (T.M.R.); (B.R.W.); (P.W.B.); (A.C.E.)
| | - Suriyan Ponnusamy
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.P.); (S.A.)
| | - Sarah Asemota
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.P.); (S.A.)
| | - Patrick W. Blackburn
- Division of Gynecologic Oncology, West Cancer Center and Research Institute, Memphis, TN 38138, USA; (M.A.U.); (T.M.R.); (B.R.W.); (P.W.B.); (A.C.E.)
| | - Yinan Wang
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Adam C. ElNaggar
- Division of Gynecologic Oncology, West Cancer Center and Research Institute, Memphis, TN 38138, USA; (M.A.U.); (T.M.R.); (B.R.W.); (P.W.B.); (A.C.E.)
| | - Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.P.); (S.A.)
- Correspondence: ; Tel.: +1-901-448-2403; Fax: +1-901-448-3910
| |
Collapse
|