1
|
Kudo K, Greer YE, Crooks DR, Yang Y, Brender JR, Yoshida T, Harrington BS, Kamdar R, Korrapati S, Shibuya Y, Henegar L, Kopp J, Fujii T, Lipkowitz S, Annunziata CM. Targeting metabolic vulnerability by combining NAMPT inhibitors and disulfiram for treatment of recurrent ovarian cancer. Cell Death Dis 2025; 16:342. [PMID: 40280967 PMCID: PMC12032209 DOI: 10.1038/s41419-025-07672-3] [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: 03/22/2024] [Revised: 04/07/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Ovarian cancer (OV) has the highest mortality rate among gynecological cancers. As OV progresses, tumor cells spread outside the ovaries to the peritoneal and abdominal cavities, forming cell clusters that float in the ascitic fluid caused by peritonitis carcinomatosa, leading to further dissemination and metastasis. These cell clusters are enriched with cancer stem cells (CSCs) which are responsible for treatment resistance, recurrence, and metastasis. Therefore, targeting CSCs is a potentially effective approach for treating OV. However, understanding how CSCs acquire treatment resistance and identifying targets against CSCs remains challenging. In this study, we demonstrate that 3D-spheroids of OV cell lines exhibit higher stemness than conventional adherent cells. Metabolomics profiling studies have revealed that 3D-spheroids maintain a high-energy state through increased glucose utilization in the citric acid cycle (TCA), efficient nucleotide phosphorylation, and elevated phosphocreatine as an energy buffer. We also found that nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme for NAD+ production, is highly expressed in OV. Furthermore, the approach based on NAMPT dependence rather than histology found NAMPT to be a potential therapeutic target against CSCs, while also serving as a prognostic indicator in OV. Moreover, we identified a previously unrecognized anti-tumor mechanism whereby disulfiram, an aldehyde dehydrogenase (ALDH) inhibitor, synergistically inhibited mitochondrial function when combined with NAMPT inhibitors - leading to cell cycle arrest in G2/M. Finally, the combination of a NAMPT inhibitor and disulfiram showed significant anti-tumor effects and extended survival in an animal model. Our findings demonstrate the potential of spheroids as a preclinical model for targeting OV CSCs and also indicate that the combination of NAMPT inhibitors and disulfiram is a promising therapeutic strategy to overcome recurrent OV.
Collapse
Affiliation(s)
- Kei Kudo
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Tohoku University School of Medicine, Miyagi, Japan
| | - Yoshimi Endo Greer
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel R Crooks
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Cancer Metabolism Facility, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ye Yang
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Cancer Metabolism Facility, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jeffrey R Brender
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Cancer Metabolism Facility, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Teruhiko Yoshida
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brittney S Harrington
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rahul Kamdar
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Soumya Korrapati
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yusuke Shibuya
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Tohoku University School of Medicine, Miyagi, Japan
| | | | - Jeffrey Kopp
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Takeo Fujii
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christina M Annunziata
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Nguyen AL, Facey COB, Boman BM. The Significance of Aldehyde Dehydrogenase 1 in Cancers. Int J Mol Sci 2024; 26:251. [PMID: 39796106 PMCID: PMC11720537 DOI: 10.3390/ijms26010251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
The goal of this paper is to discuss the role of ALDH isozymes in different cancers, review advances in ALDH1-targeting cancer therapies, and explore a mechanism that explains how ALDH expression becomes elevated during cancer development. ALDH is often overexpressed in cancer, and each isoform has a unique expression pattern and a distinct role in different cancers. The abnormal expression of ALDHs in different cancer types (breast, colorectal, lung, gastric, cervical, melanoma, prostate, and renal) is presented and correlated with patient prognosis. ALDH plays a significant role in various cellular functions, such as metabolism, oxidative stress response, detoxification, and cellular differentiation. Among the ALDH families, ALDH1 has gained considerable attention as a cancer stem cell (CSC) marker due to its significant role in the maintenance of stemness and the differentiation of stem cells (SCs), along with its involvement in tumorigenesis. A description of the cellular mechanisms and physiology of ALDH1 that underlies cancer development is provided. Moreover, current advances in ALDH1-targeting cancer therapies are discussed.
Collapse
Affiliation(s)
- Anh L. Nguyen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Caroline O. B. Facey
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Bruce M. Boman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Xu X, Han Y, Deng J, Wang S, Zhuo S, Zhao K, Zhou W. Repurposing disulfiram with CuET nanocrystals: Enhancing anti-pyroptotic effect through NLRP3 inflammasome inhibition for treating inflammatory bowel diseases. Acta Pharm Sin B 2024; 14:2698-2715. [PMID: 38828135 PMCID: PMC11143773 DOI: 10.1016/j.apsb.2024.03.003] [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: 11/15/2023] [Revised: 01/04/2024] [Accepted: 01/29/2024] [Indexed: 06/05/2024] Open
Abstract
Drug repurposing offers a valuable strategy for identifying new therapeutic applications for existing drugs. Recently, disulfiram (DSF), a drug primarily used for alcohol addiction treatment, has emerged as a potential treatment for inflammatory diseases by inhibiting pyroptosis, a form of programmed cell death. The therapeutic activity of DSF can be further enhanced by the presence of Cu2+, although the underlying mechanism of this enhancement remains unclear. In this study, we investigated the mechanistic basis of Cu2+-induced enhancement and discovered that it is attributed to the formation of a novel copper ethylthiocarbamate (CuET) complex. CuET exhibited significantly stronger anti-pyroptotic activity compared to DSF and employed a distinct mechanism of action. However, despite its potent activity, CuET suffered from poor solubility and limited permeability, as revealed by our druggability studies. To overcome these intrinsic limitations, we developed a scalable method to prepare CuET nanocrystals (CuET NCs) using a metal coordination-driven self-assembly approach. Pharmacokinetic studies demonstrated that CuET NCs exhibited a 6-fold improvement in bioavailability. Notably, CuET NCs exhibited high biodistribution in the intestine, suggesting their potential application for the treatment of inflammatory bowel diseases (IBDs). To evaluate their therapeutic efficacy in vivo, we employed a murine model of DSS-induced colitis and observed that CuET NCs effectively attenuated inflammation and ameliorated colitis symptoms. Our findings highlight the discovery of CuET as a potent anti-pyroptotic agent, and the development of CuET NCs represents a novel approach to enhance the druggability of CuET.
Collapse
Affiliation(s)
- Xueming Xu
- Hematology and Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yuanfeng Han
- Hematology and Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Jiali Deng
- Hematology and Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
- Hunan Chidren's Hospital, Changsha 410007, China
| | - Shengfeng Wang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Shijie Zhuo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Kai Zhao
- Hematology and Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha 410008, China
| |
Collapse
|
4
|
Conforti RA, Delsouc MB, Zorychta E, Telleria CM, Casais M. Copper in Gynecological Diseases. Int J Mol Sci 2023; 24:17578. [PMID: 38139406 PMCID: PMC10743751 DOI: 10.3390/ijms242417578] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Copper (Cu) is an essential micronutrient for the correct development of eukaryotic organisms. This metal plays a key role in many cellular and physiological activities, including enzymatic activity, oxygen transport, and cell signaling. Although the redox activity of Cu is crucial for enzymatic reactions, this property also makes it potentially toxic when found at high levels. Due to this dual action of Cu, highly regulated mechanisms are necessary to prevent both the deficiency and the accumulation of this metal since its dyshomeostasis may favor the development of multiple diseases, such as Menkes' and Wilson's diseases, neurodegenerative diseases, diabetes mellitus, and cancer. As the relationship between Cu and cancer has been the most studied, we analyze how this metal can affect three fundamental processes for tumor progression: cell proliferation, angiogenesis, and metastasis. Gynecological diseases are characterized by high prevalence, morbidity, and mortality, depending on the case, and mainly include benign and malignant tumors. The cellular processes that promote their progression are affected by Cu, and the mechanisms that occur may be similar. We analyze the crosstalk between Cu deregulation and gynecological diseases, focusing on therapeutic strategies derived from this metal.
Collapse
Affiliation(s)
- Rocío A. Conforti
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), San Luis CP D5700HHW, Argentina; (R.A.C.); (M.B.D.)
| | - María B. Delsouc
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), San Luis CP D5700HHW, Argentina; (R.A.C.); (M.B.D.)
| | - Edith Zorychta
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada;
| | - Carlos M. Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada;
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Marilina Casais
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), San Luis CP D5700HHW, Argentina; (R.A.C.); (M.B.D.)
| |
Collapse
|
5
|
Harrington BS, Kamdar R, Ning F, Korrapati S, Caminear MW, Hernandez LF, Butcher D, Edmondson EF, Traficante N, Hendley J, Gough M, Rogers R, Lourie R, Shetty J, Tran B, Elloumi F, Abdelmaksoud A, Nag ML, Mazan-Mamczarz K, House CD, Hooper JD, Annunziata CM. UGDH promotes tumor-initiating cells and a fibroinflammatory tumor microenvironment in ovarian cancer. J Exp Clin Cancer Res 2023; 42:270. [PMID: 37858159 PMCID: PMC10585874 DOI: 10.1186/s13046-023-02820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/02/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is a global health burden, with the poorest five-year survival rate of the gynecological malignancies due to diagnosis at advanced stage and high recurrence rate. Recurrence in EOC is driven by the survival of chemoresistant, stem-like tumor-initiating cells (TICs) that are supported by a complex extracellular matrix and immunosuppressive microenvironment. To target TICs to prevent recurrence, we identified genes critical for TIC viability from a whole genome siRNA screen. A top hit was the cancer-associated, proteoglycan subunit synthesis enzyme UDP-glucose dehydrogenase (UGDH). METHODS Immunohistochemistry was used to characterize UGDH expression in histological and molecular subtypes of EOC. EOC cell lines were subtyped according to the molecular subtypes and the functional effects of modulating UGDH expression in vitro and in vivo in C1/Mesenchymal and C4/Differentiated subtype cell lines was examined. RESULTS High UGDH expression was observed in high-grade serous ovarian cancers and a distinctive survival prognostic for UGDH expression was revealed when serous cancers were stratified by molecular subtype. High UGDH was associated with a poor prognosis in the C1/Mesenchymal subtype and low UGDH was associated with poor prognosis in the C4/Differentiated subtype. Knockdown of UGDH in the C1/mesenchymal molecular subtype reduced spheroid formation and viability and reduced the CD133 + /ALDH high TIC population. Conversely, overexpression of UGDH in the C4/Differentiated subtype reduced the TIC population. In co-culture models, UGDH expression in spheroids affected the gene expression of mesothelial cells causing changes to matrix remodeling proteins, and fibroblast collagen production. Inflammatory cytokine expression of spheroids was altered by UGDH expression. The effect of UGDH knockdown or overexpression in the C1/ Mesenchymal and C4/Differentiated subtypes respectively was tested on mouse intrabursal xenografts and showed dynamic changes to the tumor stroma. Knockdown of UGDH improved survival and reduced tumor burden in C1/Mesenchymal compared to controls. CONCLUSIONS These data show that modulation of UGDH expression in ovarian cancer reveals distinct roles for UGDH in the C1/Mesenchymal and C4/Differentiated molecular subtypes of EOC, influencing the tumor microenvironmental composition. UGDH is a strong potential therapeutic target in TICs, for the treatment of EOC, particularly in patients with the mesenchymal molecular subtype.
Collapse
Affiliation(s)
- Brittney S Harrington
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rahul Kamdar
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Franklin Ning
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Soumya Korrapati
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael W Caminear
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lidia F Hernandez
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, NCI, Frederick, MD, 21702, USA
| | - Elijah F Edmondson
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, NCI, Frederick, MD, 21702, USA
| | - Nadia Traficante
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Joy Hendley
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Madeline Gough
- Mater Brisbane Hospital, Mater Health Services, South Brisbane, QLD, 4101, Australia
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| | - Rebecca Rogers
- Mater Brisbane Hospital, Mater Health Services, South Brisbane, QLD, 4101, Australia
| | - Rohan Lourie
- Mater Brisbane Hospital, Mater Health Services, South Brisbane, QLD, 4101, Australia
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| | - Jyoti Shetty
- CCR Sequencing Facility, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Bao Tran
- CCR Sequencing Facility, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Fathi Elloumi
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Abdalla Abdelmaksoud
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Madhu Lal Nag
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Krystyna Mazan-Mamczarz
- Functional Genomics Lab, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Carrie D House
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Present address: Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - John D Hooper
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| | - Christina M Annunziata
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
Dancik GM, Varisli L, Tolan V, Vlahopoulos S. Aldehyde Dehydrogenase Genes as Prospective Actionable Targets in Acute Myeloid Leukemia. Genes (Basel) 2023; 14:1807. [PMID: 37761947 PMCID: PMC10531322 DOI: 10.3390/genes14091807] [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: 07/13/2023] [Revised: 08/29/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
It has been previously shown that the aldehyde dehydrogenase (ALDH) family member ALDH1A1 has a significant association with acute myeloid leukemia (AML) patient risk group classification and that AML cells lacking ALDH1A1 expression can be readily killed via chemotherapy. In the past, however, a redundancy between the activities of subgroup members of the ALDH family has hampered the search for conclusive evidence to address the role of specific ALDH genes. Here, we describe the bioinformatics evaluation of all nineteen member genes of the ALDH family as prospective actionable targets for the development of methods aimed to improve AML treatment. We implicate ALDH1A1 in the development of recurrent AML, and we show that from the nineteen members of the ALDH family, ALDH1A1 and ALDH2 have the strongest association with AML patient risk group classification. Furthermore, we discover that the sum of the expression values for RNA from the genes, ALDH1A1 and ALDH2, has a stronger association with AML patient risk group classification and survival than either one gene alone does. In conclusion, we identify ALDH1A1 and ALDH2 as prospective actionable targets for the treatment of AML in high-risk patients. Substances that inhibit both enzymatic activities constitute potentially effective pharmaceutics.
Collapse
Affiliation(s)
- Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey; (L.V.); (V.T.)
| | - Veysel Tolan
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey; (L.V.); (V.T.)
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| |
Collapse
|
7
|
Dancik GM, Varisli L, Vlahopoulos SA. The Molecular Context of Oxidant Stress Response in Cancer Establishes ALDH1A1 as a Critical Target: What This Means for Acute Myeloid Leukemia. Int J Mol Sci 2023; 24:ijms24119372. [PMID: 37298333 DOI: 10.3390/ijms24119372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The protein family of aldehyde dehydrogenases (ALDH) encompasses nineteen members. The ALDH1 subfamily consists of enzymes with similar activity, having the capacity to neutralize lipid peroxidation products and to generate retinoic acid; however, only ALDH1A1 emerges as a significant risk factor in acute myeloid leukemia. Not only is the gene ALDH1A1 on average significantly overexpressed in the poor prognosis group at the RNA level, but its protein product, ALDH1A1 protects acute myeloid leukemia cells from lipid peroxidation byproducts. This capacity to protect cells can be ascribed to the stability of the enzyme under conditions of oxidant stress. The capacity to protect cells is evident both in vitro, as well as in mouse xenografts of those cells, shielding cells effectively from a number of potent antineoplastic agents. However, the role of ALDH1A1 in acute myeloid leukemia has been unclear in the past due to evidence that normal cells often have higher aldehyde dehydrogenase activity than leukemic cells. This being true, ALDH1A1 RNA expression is significantly associated with poor prognosis. It is hence imperative that ALDH1A1 is methodically targeted, particularly for the acute myeloid leukemia patients of the poor prognosis risk group that overexpress ALDH1A1 RNA.
Collapse
Affiliation(s)
- Garrett M Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Spiros A Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
| |
Collapse
|
8
|
Kamdar RD, Harrington BS, Attar E, Korrapati S, Shetty J, Zhao Y, Tran B, Wong N, House CD, Annunziata CM. NF-κB Signaling Modulates miR-452-5p and miR-335-5p Expression to Functionally Decrease Epithelial Ovarian Cancer Progression in Tumor-Initiating Cells. Int J Mol Sci 2023; 24:7826. [PMID: 37175530 PMCID: PMC10178396 DOI: 10.3390/ijms24097826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Epithelial ovarian cancer (EOC) remains the fifth leading cause of cancer-related death in women worldwide, partly due to the survival of chemoresistant, stem-like tumor-initiating cells (TICs) that promote disease relapse. We previously described a role for the NF-κB pathway in promoting TIC chemoresistance and survival through NF-κB transcription factors (TFs) RelA and RelB, which regulate genes important for the inflammatory response and those associated with cancer, including microRNAs (miRNAs). We hypothesized that NF-κB signaling differentially regulates miRNA expression through RelA and RelB to support TIC persistence. Inducible shRNA was stably expressed in OV90 cells to knockdown RELA or RELB; miR-seq analyses identified differentially expressed miRNAs hsa-miR-452-5p and hsa-miR-335-5p in cells grown in TIC versus adherent conditions. We validated the miR-seq findings via qPCR in TIC or adherent conditions with RELA or RELB knocked-down. We confirmed decreased expression of hsa-miR-452-5p when either RELA or RELB were depleted and increased expression of hsa-miR-335-5p when RELA was depleted. Either inhibiting miR-452-5p or mimicking miR-335-5p functionally decreased the stem-like potential of the TICs. These results highlight a novel role of NF-κB TFs in modulating miRNA expression in EOC cells, thus opening a better understanding toward preventing recurrence of EOC.
Collapse
Affiliation(s)
- Rahul D. Kamdar
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brittney S. Harrington
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emma Attar
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Soumya Korrapati
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jyoti Shetty
- CCR Sequencing Facility, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Yongmei Zhao
- CCR Sequencing Facility, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Bao Tran
- CCR Sequencing Facility, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Nathan Wong
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Carrie D. House
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christina M. Annunziata
- Women’s Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
9
|
Wang Q, Li Z, Hao Y, Zhang Y, Zhang C. Near-Infrared Fluorescence Probe with a New Recognition Moiety for Specific Detection and Imaging of Aldehyde Dehydrogenase Expecting the Identification and Isolation of Cancer Stem Cells. Anal Chem 2022; 94:17328-17333. [PMID: 36453832 DOI: 10.1021/acs.analchem.2c04801] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Aldehyde dehydrogenase (ALDH) is a vital enzyme that converts aldehyde to acetic acid during alcohol metabolism. ALDH is also a cellular marker of cancer stem cells (CSCs), which plays an important role in cancer diagnosis and prognosis assessment. Therefore, there is a need to explore convenient, selective, and sensitive methods for the detection and imaging of ALDH. Because of the low background fluorescence and high penetration, near-infrared (NIR) fluorescent probes are powerful tools for the detection of ALDH. Until now, only one NIR fluorescent probe has been reported for detecting ALDH. Hence, we synthesized a novel NIR fluorescent probe, Probe-ALDH, by linking the new specific recognition moiety 4-hydroxymethyl benzaldehyde with NIR fluorophore AXPI. Compared with the existing ALDH fluorescent probes, Probe-ALDH has excellent properties, such as a new specific recognition moiety without the substitution of benzaldehyde, a simple synthesis method, emission wavelength in the NIR region, reaction time of only 30 min, and a detection limit as low as 0.03 U·mL-1, which is better than those of the previously reported probes. The probe effectively eliminates the interference from reactive oxygen species (ROS), amino acids, and amines. More importantly, the flow cytometry results showed that Probe-ALDH has great potential applications in the identification and isolation of CSCs. Ultimately, it was successfully applied to the imaging analysis of endogenous ALDH in HepG2 cells by the addition of inhibitor disulfiram. The excellent performance of Probe-ALDH makes it a promising candidate for drug discovery, cancer diagnosis, and so forth.
Collapse
Affiliation(s)
- Qiuyue Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Zhao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Yitong Hao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Yuan Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Chengxiao Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China
| |
Collapse
|
10
|
Konishi I, Abiko K, Hayashi T, Yamanoi K, Murakami R, Yamaguchi K, Hamanishi J, Baba T, Matsumura N, Mandai M. Peritoneal dissemination of high-grade serous ovarian cancer: pivotal roles of chromosomal instability and epigenetic dynamics. J Gynecol Oncol 2022; 33:e83. [PMID: 36032027 PMCID: PMC9428305 DOI: 10.3802/jgo.2022.33.e83] [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: 06/02/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Epithelial ovarian cancer remains the lethal gynecological malignancy in women. The representative histotype is high-grade serous carcinoma (HGSC), and most patients with HGSC present at advanced stages with peritoneal dissemination. Since the peritoneal dissemination is the most important factor for poor prognosis of the patients, complete exploration for its molecular mechanisms is mandatory. In this narrative review, being based on the clinical, pathologic, and genomic findings of HGSC, chromosomal instability and epigenetic dynamics have been discussed as the potential drivers for cancer development in the fallopian tube, acquisition of cancer stem cell (CSC)-like properties, and peritoneal metastasis of HGSC. The natural history of carcinogenesis with clonal evolution, and adaptation to microenvironment of peritoneal dissemination of HGSC should be targeted in the novel development of strategies for prevention, early detection, and precision treatment for patients with HGSC.
Collapse
Affiliation(s)
- Ikuo Konishi
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.,Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.,Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takuma Hayashi
- Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
11
|
Muralikrishnan V, Fang F, Given TC, Podicheti R, Chtcherbinine M, Metcalfe TX, Sriramkumar S, O’Hagan HM, Hurley TD, Nephew KP. A Novel ALDH1A1 Inhibitor Blocks Platinum-Induced Senescence and Stemness in Ovarian Cancer. Cancers (Basel) 2022; 14:3437. [PMID: 35884498 PMCID: PMC9318275 DOI: 10.3390/cancers14143437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is a deadly disease attributed to late-stage detection as well as recurrence and the development of chemoresistance. Ovarian cancer stem cells (OCSCs) are hypothesized to be largely responsible for the emergence of chemoresistant tumors. Although chemotherapy may initially succeed at decreasing the size and number of tumors, it leaves behind residual malignant OCSCs. In this study, we demonstrate that aldehyde dehydrogenase 1A1 (ALDH1A1) is essential for the survival of OCSCs. We identified a first-in-class ALDH1A1 inhibitor, compound 974, and used 974 as a tool to decipher the mechanism of stemness regulation by ALDH1A1. The treatment of OCSCs with 974 significantly inhibited ALDH activity, the expression of stemness genes, and spheroid and colony formation. An in vivo limiting dilution assay demonstrated that 974 significantly inhibited CSC frequency. A transcriptomic sequencing of cells treated with 974 revealed a significant downregulation of genes related to stemness and chemoresistance as well as senescence and the senescence-associated secretory phenotype (SASP). We confirmed that 974 inhibited the senescence and stemness induced by platinum-based chemotherapy in functional assays. Overall, these data establish that ALDH1A1 is essential for OCSC survival and that ALDH1A1 inhibition suppresses chemotherapy-induced senescence and stemness. Targeting ALDH1A1 using small-molecule inhibitors in combination with chemotherapy therefore presents a promising strategy to prevent ovarian cancer recurrence and has the potential for clinical translation.
Collapse
Affiliation(s)
- Vaishnavi Muralikrishnan
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Fang Fang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Tyler C. Given
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 46202, USA;
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Tara X. Metcalfe
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Shruthi Sriramkumar
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
| | - Heather M. O’Hagan
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Thomas D. Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Kenneth P. Nephew
- Cell, Molecular and Cancer Biology Graduate Program, Medical Sciences Department, Indiana University School of Medicine, Bloomington, IN 47405, USA; (V.M.); (T.C.G.); (T.X.M.); (S.S.); (H.M.O.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Anatomy, Cell Biology and Physiology, Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|