1
|
Marchetti M, Gros BP, Muller O, Ferlay C, Ltaief Z, Kirsch M, Nowacka A, Pascale P, Meyer P, Yerly P, Monney P, Antiochos P, Skali H, Cikes M, Hullin R, Lu H. Impact of Digoxin Utilization on Gastrointestinal Bleeding in Patients With Continuous-Flow Left Ventricular Assist Devices: A Systematic Review and Meta-Analysis. ASAIO J 2025:00002480-990000000-00704. [PMID: 40382704 DOI: 10.1097/mat.0000000000002473] [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: 05/20/2025] Open
Abstract
Continuous-flow left ventricular assist devices (CF-LVADs) improve quality of life and survival in patients with advanced heart failure but are frequently complicated by gastrointestinal bleeding (GIB). Reduced pulsatile flow may induce mucosal hypoxia, upregulating factors such as hypoxia-inducible factor (HIF)-1α and triggering neo-angiogenesis, leading to the development of gastrointestinal angiodysplasias (GIADs), a common cause of GIB. Digoxin inhibits HIF-1α and may prevent GIAD development, although its impact on the incidence of GIB remains uncertain. This meta-analysis (PROSPERO ID: CRD42024626222) evaluated the association between digoxin use and GIB occurrence (primary outcome) in patients with CF-LVADs. Research articles including adults with CF-LVADs, comparing digoxin users versus nonusers were included. Overall, four studies were included (n = 14,917; age 55 ± 13 years, 21% female) with 2,742 patients in the digoxin group and 12,175 in the no-digoxin group. Continuous-flow left ventricular assist device was axial (HeartMate II) in 78% of cases and centrifugal (HeartMate 3/HeartWare) in 22%. Digoxin use was associated with a nonsignificant lower risk of GIB (hazard ratio [HR]: 0.70; 95% confidence interval [CI]: 0.49-1.01). However, regarding GIAD-related GIB, digoxin was associated with a significantly lower risk (HR: 0.33; 95% CI: 0.13-0.82). Among 14,917 patients with CF-LVADs, digoxin use was associated with a trend toward a lower risk of GIB and a lower risk of GIAD-related GIB.
Collapse
Affiliation(s)
- Matteo Marchetti
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Barbara Pitta Gros
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Olivier Muller
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Clémence Ferlay
- Division of Adult Intensive Care Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Zied Ltaief
- Division of Adult Intensive Care Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Matthias Kirsch
- Division of Cardiac Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Anna Nowacka
- Division of Cardiac Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Patrizio Pascale
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philippe Meyer
- Division of Cardiology, Geneva University Hospital, University of Geneva, Geneva, Switzerland
| | - Patrick Yerly
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Pierre Monney
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Panagiotis Antiochos
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Hicham Skali
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maja Cikes
- Department of Cardiovascular Diseases, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Roger Hullin
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Henri Lu
- From the Division of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
2
|
Li WJ, Chen YC, Lin YA, Zou YQ, Hu GS, Yang JJ, Nie XY, Li MY, Wang YR, He YH, Zhao Y, Tan YH, Deng X, He WL, Cheng Y, Fu FM, Liu W. Hypoxia-induced PRMT1 methylates HIF2β to promote breast tumorigenesis via enhancing glycolytic gene transcription. Cell Rep 2025; 44:115487. [PMID: 40173041 DOI: 10.1016/j.celrep.2025.115487] [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: 07/17/2024] [Revised: 01/28/2025] [Accepted: 03/07/2025] [Indexed: 04/04/2025] Open
Abstract
Hypoxia-induced metabolic reprogramming is closely linked to breast cancer progression. Through transcriptomic analysis, we identified PRMT1 as a direct target of hypoxia-inducible factor 1α (HIF1α) under hypoxic conditions in breast cancer cells. In turn, PRMT1 enhances the expression of HIF1α-driven glycolytic genes. Mechanistically, PRMT1 methylates HIF2β at arginine 42, facilitating the formation, chromatin binding, and the transcriptional activity of the HIF1α/HIF2β heterodimer. Genetic and pharmacological inhibition of PRMT1 suppresses HIF2β methylation, HIF1α/HIF2β heterodimer formation, chromatin binding, glycolytic gene expression, lactate production, and the malignant behaviors of breast cancer cells. Moreover, combination treatment with iPRMT1, a PRMT1 inhibitor, and menadione, an HIF1α/P300 interaction inhibitor, demonstrates synergistic effects in suppressing breast tumor growth. Clinically, PRMT1 and PRMT1-mediated HIF2β methylation were significantly elevated in breast tumors compared with adjacent normal tissues. In conclusion, our findings reveal the critical role of PRMT1-mediated arginine methylation in glycolytic gene expression, metabolic reprogramming, and breast tumor growth.
Collapse
Affiliation(s)
- Wen-Juan Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan-Chao Chen
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yi-An Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yi-Qin Zou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Guo-Sheng Hu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jing-Jing Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xin-Yu Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Mei-Yan Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yi-Ran Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yao-Hui He
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yu-Hua Tan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wei-Ling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Fang-Meng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| | - Wen Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
3
|
Weng L, Zhao M, Chen Z, Zhu L. Hypoxia-Targeted Responsive Delivery of Doxorubicin and Digoxin for Synergistic Treatment of Triple-Negative Breast Cancer. Mol Pharm 2025; 22:2142-2158. [PMID: 40059340 DOI: 10.1021/acs.molpharmaceut.4c01325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2025]
Abstract
To enhance the therapeutic efficacy and safety of triple-negative breast cancer (TNBC) treatment, we developed a hypoxia-responsive drug delivery system utilizing digoxin (DIG) to inhibit HIF-1α and sensitize TNBC to doxorubicin (DOX). DIG, a cardiac steroid with a well-characterized pharmacological mechanism, was encapsulated in micelles composed of methoxy-polyethylene glycol (mPEG) and poly(lactic acid) (PLA) copolymers, incorporating an azobenzene (AZO) trigger for hypoxia-sensitive drug release. The loading ratio of DOX to DIG was optimized based on DIG's minimum effective dose. In vitro and in vivo studies demonstrated that the micelles efficiently delivered their payload to hypoxic tumor regions, enabling rapid drug release. DIG-mediated HIF-1α inhibition enhanced TNBC sensitivity to DOX, leading to significant suppression of both primary tumor growth and pulmonary metastasis. This study presents a promising and clinically feasible strategy for TNBC and other hypoxia-driven malignancies.
Collapse
Affiliation(s)
- Lingyan Weng
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| | - Min Zhao
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| | - Zhongping Chen
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| | - Li Zhu
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| |
Collapse
|
4
|
Karimnia N, Wilson AL, Doran BR, Do J, Matthews A, Ho GY, Plebanski M, Jobling TW, Stephens AN, Bilandzic M. A Novel 3D High-Throughput Phenotypic Drug Screening Pipeline to Identify Drugs with Repurposing Potential for the Treatment of Ovarian Cancer. Adv Healthc Mater 2025; 14:e2404117. [PMID: 40109101 PMCID: PMC12023816 DOI: 10.1002/adhm.202404117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Ovarian cancer (OC) poses a significant clinical challenge due to its high recurrence rates and resistance to standard therapies, particularly in advanced stages where recurrence is common, and treatment is predominantly palliative. Personalized treatments, while effective in other cancers, remain underutilized in OC due to a lack of reliable biomarkers predicting clinical outcomes. Accordingly, precision medicine approaches are limited, with PARP inhibitors showing efficacy only in specific genetic contexts. Drug repurposing offers a promising, rapidly translatable strategy by leveraging existing pharmacological data to identify new treatments for OC. Patient-derived polyclonal spheroids, isolated from ascites fluid closely mimic the clinical behavior of OC, providing a valuable model for drug testing. Using these spheroids, a high-throughput drug screening pipeline capable of evaluating both cytotoxicity and anti-migratory properties of a diverse drug library, including FDA-approved, investigational, and newly approved compounds is developed. The findings highlight the importance of 3D culture systems, revealing a poor correlation between drug efficacy in traditional 2D models and more clinically relevant 3D spheroids. This approach has expedited the identification of promising candidates, such as rapamycin, which demonstrated limited activity as a monotherapy but synergized effectively with standard treatments like cisplatin and paclitaxel in vitro. In combination with platinum-based therapy, Rapamycin led to significant in vitro cytotoxicity and a marked reduction in tumor burden in a syngeneic in vivo model. This proof-of-concept study underscores the potential of drug repurposing to rapidly advance new treatments into clinical trials for OC, offering renewed hope for patients with advanced disease.
Collapse
Affiliation(s)
- Nazanin Karimnia
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Amy L. Wilson
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Brittany R. Doran
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Jennie Do
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Amelia Matthews
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Gwo Y. Ho
- School of Clinical SciencesMonash University27 Rainforest WalkClaytonVIC3168Australia
- Department of OncologyMonash Health823–865 Centre RdBentleighVIC3165Australia
| | - Magdalena Plebanski
- School of Health and Biomedical SciencesRMIT UniversityBundoora campus West203, 1/2 Plenty RdBundooraVIC3083Australia
| | - Thomas W. Jobling
- Monash Medical CentreDepartment of Gynecology OncologyMonash Health823–865 Centre RdBentleigh EastVIC3165Australia
| | - Andrew N. Stephens
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| |
Collapse
|
5
|
Eghbalifard N, Nouri N, Rouzbahani S, Bakhshi M, Ghasemi Kahrizsangi N, Golafshan F, Abbasi F. Hypoxia signaling in cancer: HIF-1α stimulated by COVID-19 can lead to cancer progression and chemo-resistance in oral squamous cell carcinoma (OSCC). Discov Oncol 2025; 16:399. [PMID: 40138101 PMCID: PMC11947373 DOI: 10.1007/s12672-025-02150-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
The potential implications of Coronavirus disease-2019 (COVID-19) on oral squamous cell carcinoma (OSCC) development, chemo-resistance, tumor recurrence, and patient outcomes are explored, emphasizing the urgent need for tailored therapeutic strategies to mitigate these risks. The role of hypoxia-inducible factor 1-alpha (HIF-1α) in OSCC studies has highlighted HIF-1α as a crucial prognostic marker in OSCC, with implications for disease prognosis and patient survival. Its overexpression has been linked to aggressive subtypes in early OSCC stages, indicating its significance as an early biomarker for disease progression. Moreover, dysplastic lesions with heightened HIF-1α expression exhibit a greater propensity for malignant transformation, underscoring its role in early oral carcinogenesis. Cancer patients, including those with OSCC, face an elevated risk of severe COVID-19 complications, which may further impact cancer progression and treatment outcomes. Understanding the interplay between COVID-19 infection, HIF-1α activation, and OSCC pathogenesis is crucial for enhancing clinical management strategies. So, insights from this review shed light on the significance of HIF-1α in OSCC tumorigenesis, metastasis formation, and patient prognosis. The review underscores the need for further research to elucidate the precise mechanisms through which HIF-1α modulates cancer progression and chemo-resistance in the context of COVID-19 infection. Such knowledge is essential for developing targeted therapeutic interventions to improve outcomes for OSCC patients.
Collapse
Affiliation(s)
- Negar Eghbalifard
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nikta Nouri
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shiva Rouzbahani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Bakhshi
- Islamic Azad University of Najaf Abad, Affiliated Hospitals, Isfahan, Iran
| | - Negin Ghasemi Kahrizsangi
- Child Growth and Development Research Center, Research Institute for Primary Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Faraz Golafshan
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Abbasi
- Department of Obstetrics and Gynecology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran.
| |
Collapse
|
6
|
Poluektov YM, Lopina OD, Strelkova MA, Kuleshova ID, Makarov AA, Petrushanko IY. Mechanisms mediating effects of cardiotonic steroids in mammalian blood cells. Front Pharmacol 2025; 16:1520927. [PMID: 40196366 PMCID: PMC11973394 DOI: 10.3389/fphar.2025.1520927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Cardiotonic steroids (CTSs) were known as steroidal plant compounds that exert cellular effects by the binding to Na,K-ATPase. Earlier, plant (exogenous) CTSs were used to treat chronic heart failure. By now, endogenous CTS have been identified in mammals, and their concentrations in the blood, normally in a subnanomolar range, are altered in numerous pathologies. This indicates their role as endogenous regulators of physiological processes. CTS transport occurs primarily in the blood, yet the CTS effects on blood cells remain poorly understood. This review summarizes the CTS effects on blood cells of animals and humans under normal and pathological conditions, and analyzes their action based on known mechanisms of action in mammalian cells. At high concentrations (greater than 10-9 M), CTS binding to Na,K-ATPase inhibits the enzyme, whereas lower concentrations of CTSs induce signaling cascades or activate the enzyme. All these mechanisms are shown to be present in blood cells. The particular CTS effect is determined by the CTS type, its concentration, the isoform composition of the catalytic α-subunit of Na,K-ATPase in the cell, and other cell features. It has been demonstrated that all blood cell types (erythrocytes, leukocytes, and platelets) expressed both ubiquitously distributed α1-isoform and tissue-specific α3-subunit, which exhibits a different ion and CTS affinity compared to α1. This results in a wide spectrum of blood cell responses to fluctuations in CTS levels in the blood. In particular, an increase in the level of endogenous CTSs by a more twofold is sufficient to induce a decline in the activity of erythrocyte Na,K-ATPase. The administration of exogenous CTSs is able to modulate the proinflammatory activity of leukocytes, which is attributed to the activation of signaling cascades, and to exert an influence on platelet activation. Hence, alterations of CTS levels in bloodstream significantly affect the functionality of blood cells, contributing to the organism's adaptive response. On top of this, a comparison of the effects of CTSs on human leukocytes and rodent leukocytes carrying the CTS-resistant α1-isoform often reveals opposite effects, thus indicating that rodents are an unsuitable model for studying CTS effects on these cells.
Collapse
Affiliation(s)
- Yuri M. Poluektov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Olga D. Lopina
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Strelkova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Iuliia D. Kuleshova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Irina Yu. Petrushanko
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
7
|
Rahman MA, Jalouli M, Bhajan SK, Al-Zharani M, Harrath AH. The Role of Hypoxia-Inducible Factor-1α (HIF-1α) in the Progression of Ovarian Cancer: Perspectives on Female Infertility. Cells 2025; 14:437. [PMID: 40136686 PMCID: PMC11941611 DOI: 10.3390/cells14060437] [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: 02/09/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Hypoxia-Inducible Factor-1α (HIF-1α) is crucial in the progression of ovarian cancer, especially in influencing its tumor microenvironment and promoting pathogenic pathways that worsen female infertility. In hypoxic settings, HIF-1α is stabilized and activates the transcription of genes associated with angiogenesis, metabolic reprogramming, epithelial-to-mesenchymal transition, and therapeutic resistance. Angiogenesis and glycolytic reprogramming mediated by HIF-1 tumor proliferation, survival, and metastasis. Its dysfunction concurrently impairs ovarian homeostasis, undermining follicular growth, hormone synthesis, and the ovarian vascular network, consequently contributing to infertility. Moreover, HIF-1α induces persistent inflammation and oxidative stress, promoting an environment damaging to reproductive health. Due to its dual function in ovarian cancer growth and infertility, HIF-1α is a potential therapeutic target. Strategies including small molecule inhibitors and nanoparticle-mediated delivery of drugs possess the potential to reduce HIF-1α activity, hence reducing cancer progression while protecting fertility. This review seeks to clarify the molecular basis of HIF-1α in ovarian cancer and its effects on female infertility, providing insights into novel treatment approaches that target both controlling the disease and preserving fertility.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA;
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia; (M.J.); (M.A.-Z.)
| | - Sujay Kumar Bhajan
- Department of Biotechnology and Genetic Engineering, Faculty of Life Sciences, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh;
| | - Mohammed Al-Zharani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia; (M.J.); (M.A.-Z.)
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
8
|
Liu H, Huang C, Liu Z, Li Y, Zhu Y, Gao M, Chen J, Zhang H, Xiao Z, Zhao W. Systematic drug screening and target analysis identify digitoxin as a potential therapy for uveal melanoma. Br J Pharmacol 2025; 182:1275-1291. [PMID: 39617007 DOI: 10.1111/bph.17405] [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: 07/06/2024] [Revised: 10/11/2024] [Accepted: 10/19/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Cardiac glycosides (CGs), traditionally prescribed for heart failure and arrhythmias, show anticancer potential. However, their mechanisms for preferential inhibition of tumour tissue and constituent malignant cells are not fully elucidated. This study aims to elucidate the therapeutic benefits of CGs in targeting specific tumours and dissect their multi-targeting mechanisms that confer their cytotoxicity against malignant cells. EXPERIMENTAL APPROACH We designed an integrated workflow to identify therapeutic CGs with high toxicity to certain cancers, investigating their multi-target effects, assessing their toxicity to malignant cells and analysing the prognostic relevance of CGs' target genes. The computational findings were confirmed through gene knockdown, cell viability assays, reactive oxygen species (ROS) measurements and so forth. KEY RESULTS CGs modulate multiple genes crucial for ion homeostasis, oxidative stress and apoptosis, with a particularly strong inhibitory effects on uveal melanoma (UVM). Notably, digitoxin suppresses UVM cell proliferation and induces ROS levels by simultaneously targeting STAT3 and KLF5. Single-cell transcriptomic analysis revealed that malignant cells are likely more vulnerable to CGs due to their higher expression of CG target genes compared with surrounding cells in the UVM microenvironment. CONCLUSIONS AND IMPLICATIONS Given UVM's limited options, our study highlights the potential of digitoxin as a promising novel therapeutic agent for this aggressive and rare ocular cancer. Our comprehensive approach is effective in identifying the potent, cancer-specific therapeutic agents from herbal plants.
Collapse
Affiliation(s)
- Huilin Liu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Chao Huang
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Zhenni Liu
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Yuhan Li
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Yanan Zhu
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Min Gao
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Jing Chen
- Department of Obstetrics, Xi'an New Chang'an Maternity Hospital, Xi'an, Shaanxi, China
- Shaanxi Stem Cell Engineering Application Research Center, Shaanxi Jiuzhou Biomedical Science and Technology Group, Xi'an, Shaanxi, China
| | - Hui Zhang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, China
| | - Zhengtao Xiao
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
9
|
Jalouli M. Emerging Role of Hypoxia-Inducible Factors (HIFs) in Modulating Autophagy: Perspectives on Cancer Therapy. Int J Mol Sci 2025; 26:1752. [PMID: 40004215 PMCID: PMC11855875 DOI: 10.3390/ijms26041752] [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: 12/27/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are master regulators of cellular responses to low oxygen levels and modulate autophagy, a conserved process essential for maintaining homeostasis. Under hypoxic conditions, HIFs regulate the expression of autophagy-related genes and influence autophagic flux and cellular stress responses. Dysregulated hypoxia-induced autophagy promotes cancer cell survival, metabolism, and metastasis, thereby contributing to treatment resistance. Targeting HIF-mediated pathways or modulating autophagic processes offers the potential to improve traditional cancer therapies and overcome drug resistance. Pharmacological inhibitors of HIFs or autophagy, either alone or in combination with other treatments, may disrupt the pro-survival mechanisms within the hypoxic tumor microenvironment. Further research is needed to elucidate the intricate interplay between HIF signaling and the autophagy machinery in cancer cells. Understanding these processes could pave the way for novel therapeutic strategies to enhance treatment outcomes and combat drug resistance. This review highlights the complex relationship between HIFs and autophagy in cancer development and therapy, offering insights into how targeting these pathways may improve patient outcomes.
Collapse
Affiliation(s)
- Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| |
Collapse
|
10
|
Kaewpaeng N, Wongwan S, Pansooksan K, Chailom C, Chaisupasakul P, Pekthong D, Nuengchamnong N, Jiang ZH, Bai LP, Srisawang P, Parhira S. HPLC-MS standardization and validation methods for determination of calactin content in dichloromethane fraction of Calotropis gigantea (L.) Dryand. ( Niu jiao gua) stem bark and its application in prediction of anticancer activities. Heliyon 2025; 11:e41963. [PMID: 39916826 PMCID: PMC11795781 DOI: 10.1016/j.heliyon.2025.e41963] [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: 05/16/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
Calactin, a doubly-linked cardenolide, is commonly found in Apocynaceae family including Calotropis gigantea (L.) Dryand. (Niu jiao gua, C. gigantea). This phytochemical has gained recognition for its potential as an anticancer agent and a marker for anticancer properties. Our previous reports demonstrated the remarkable anti-liver and anti-colon cancer activities both in vitro and in vivo of the dichloromethane fraction from the stem barks of C. gigantea (CGD). However, quantitative analysis of calactin content in CGD by using a high-performance liquid chromatography (HPLC) - ultraviolet (UV) method was limited. Therefore, this study aimed to develop more sensitive and reliable method for measurement of calactin in CGD by using a HPLC- electrospray ionization mass spectrometry (ESI-MS) for its application on quality control and prediction of anticancer activity of CGD. The study utilized a HPLC-ESI-MS system, negative mode, for standardization and validation. An octadecylsilane column and a mixture of acetonitrile and water containing 0.1 % formic acid in gradient system were used as a stationary phase and a mobile phase, respectively. The method was validated in terms of linearity, accuracy, precision, recovery, limit of detection (LOD), and limit of quantitation (LOQ). The linearity of the developed technique was verified within the calactin concentrations range from 1 to 50 μg/mL, exhibiting a linear coefficient of determination (R2) greater than 0.998. The LOD and LOQ were 0.1 and 1 μg/mL, respectively. The proposed methodology met the acceptance criteria according to the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) guideline Q2(R1). Therefore, the method was suitable for the quality control of CGD. Additionally, we suggest applying the established protocol to quantify the calactin contents in CGDs collected over a period of one year (12 months) to investigate the correlation to their cytotoxicities against human cell lines derived from colon cancer (HCT116), human hepatoblastoma (HepG2) and human cell line derived from stomach cancer metastasized to liver (MKN74). This will help in predicting its cytotoxicity against HCT116, HepG2 and MKN74. In summary, the established HPLC-ESI-MS method is suitable for both quality control and predicting of CGD anticancer activities for further utilization.
Collapse
Affiliation(s)
- Naphat Kaewpaeng
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, 65000, Phitsanulok, Thailand
| | - Sudaporn Wongwan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
| | - Khemmachat Pansooksan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, 65000, Phitsanulok, Thailand
| | - Chanakan Chailom
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, 65000, Phitsanulok, Thailand
| | - Pattaraporn Chaisupasakul
- Center of Excellence for Innovation in Chemistry, Naresuan University, 65000, Phitsanulok, Thailand
- Department of Physiology, Faculty of Medical Science, Naresuan University, 65000, Phitsanulok, Thailand
| | - Dumrongsak Pekthong
- Center of Excellence for Innovation in Chemistry, Naresuan University, 65000, Phitsanulok, Thailand
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
| | | | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Li-Ping Bai
- State Key Laboratory of Quality Research in Chinese Medicine, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Piyarat Srisawang
- Center of Excellence for Innovation in Chemistry, Naresuan University, 65000, Phitsanulok, Thailand
- Department of Physiology, Faculty of Medical Science, Naresuan University, 65000, Phitsanulok, Thailand
- Center of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, 65000, Phitsanulok, Thailand
| | - Supawadee Parhira
- Center of Excellence for Innovation in Chemistry, Naresuan University, 65000, Phitsanulok, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Naresuan University, 65000, Phitsanulok, Thailand
| |
Collapse
|
11
|
Wang L, Nakamura A. Where are we in targeting hypoxia-induced pathways in inflammatory arthritis? Current understanding, insights, and future directions. Int Immunopharmacol 2025; 146:113883. [PMID: 39718060 DOI: 10.1016/j.intimp.2024.113883] [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: 09/30/2024] [Revised: 11/30/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Joint tissues affected by inflammatory arthritis (IA) create hypoxic microenvironments that sustain the inflammatory response. Although targeting molecules in hypoxia-induced pathways has provided valuable insights into potential novel therapies for various types of IA, progress remains preclinical, and no clinical trials have been conducted for IA. METHODS A literature search was conducted to create a narrative review exploring the role of hypoxia and its signaling pathways in IA pathogenesis, as well as the potential and future directions for IA therapies that target hypoxia-induced molecules before moving forward to clinical applications. RESULTS Hypoxia is a prevalent feature of the IA synovial microenvironment and contributes to disease progression. Various studies and preclinical models demonstrate how hypoxia-inducible factors, vascular endothelial growth factors, and matrix metalloproteinases, among other molecules, influence rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis, and juvenile idiopathic arthritis. Despite these findings, drug development targeting these molecules in IA has been limited due to challenges in delineating the mechanistic pathways of hypoxia, the distinct roles of hypoxia-induced molecules depending on anatomical sites, and concerns regarding pharmacokinetics and patient safety. However, given that hypoxia-induced molecule-targeting therapies have been successfully approved for treating cancers and cardiovascular diseases, further research is needed to advance the application of similar medications in IA. CONCLUSIONS Given the pathogenic effects of hypoxic microenvironments in IA, it is imperative to continue gathering compelling evidence to advance hypoxia-induced therapies. Furthermore, elucidating the safety and efficacy of such drugs in various preclinical models, in collaboration with chemists and the pharmaceutical industry, is crucial for accelerating the development of novel, optimized treatment methods.
Collapse
Affiliation(s)
- Lisa Wang
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada.
| | - Akihiro Nakamura
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada; Translational Institute of Medicine, Department of Medicine, Queen's University, Ontario, Canada; Rheumatology Clinic, Kingston Health Science Centre, Kingston, Ontario, Canada.
| |
Collapse
|
12
|
Zhang J, Yao M, Xia S, Zeng F, Liu Q. Systematic and comprehensive insights into HIF-1 stabilization under normoxic conditions: implications for cellular adaptation and therapeutic strategies in cancer. Cell Mol Biol Lett 2025; 30:2. [PMID: 39757165 DOI: 10.1186/s11658-024-00682-7] [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: 09/27/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are essential transcription factors that orchestrate cellular responses to oxygen deprivation. HIF-1α, as an unstable subunit of HIF-1, is usually hydroxylated by prolyl hydroxylase domain enzymes under normoxic conditions, leading to ubiquitination and proteasomal degradation, thereby keeping low levels. Instead of hypoxia, sometimes even in normoxia, HIF-1α translocates into the nucleus, dimerizes with HIF-1β to generate HIF-1, and then activates genes involved in adaptive responses such as angiogenesis, metabolic reprogramming, and cellular survival, which presents new challenges and insights into its role in cellular processes. Thus, the review delves into the mechanisms by which HIF-1 maintains its stability under normoxia including but not limited to giving insights into transcriptional, translational, as well as posttranslational regulation to underscore the pivotal role of HIF-1 in cellular adaptation and malignancy. Moreover, HIF-1 is extensively involved in cancer and cardiovascular diseases and potentially serves as a bridge between them. An overview of HIF-1-related drugs that are approved or in clinical trials is summarized, highlighting their potential capacity for targeting HIF-1 in cancer and cardiovascular toxicity related to cancer treatment. The review provides a comprehensive insight into HIF-1's regulatory mechanism and paves the way for future research and therapeutic development.
Collapse
Affiliation(s)
- Jiayi Zhang
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Mingxuan Yao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shiting Xia
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
13
|
Ren Y, Anderson AT, Meyer G, Lauber KM, Gallucci JC, Douglas Kinghorn A. Digoxin and its Na +/K +-ATPase-targeted actions on cardiovascular diseases and cancer. Bioorg Med Chem 2024; 114:117939. [PMID: 39396465 PMCID: PMC11527570 DOI: 10.1016/j.bmc.2024.117939] [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: 08/16/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Na+/K+-ATPase (NKA) is a plasma membrane ion-transporting protein involved in the generation and maintenance of Na+ and K+ gradients across the cell membrane, which can produce a driving force for the secondary transport of metabolic substrates. NKA also regulates intracellular calcium that is responsible for modulating numerous cellular processes, while it interacts with many other proteins and functions as a signal transducer, with several signaling pathways being involved. Thus, NKA has become an important target for the treatment of human diseases. Cardiac glycosides are well-known NKA inhibitors, of which (+)-digoxin or digoxin has been long used for the treatment of congestive heart failure. Also, digoxin has exhibited potential antitumor activity, by targeting directly HIF-1α, NKA, and NF-κB. Thus, the function of NKA in human cardiovascular diseases and cancer and the therapeutic effects of digoxin on these diseases are summarized in the present review, with the correlations among digoxin, NKA, cardiovascular diseases, and cancer being discussed. Presented herein are also the antitumor potential of monosaccharide cardiac glycoside analogues of digoxin, including (-)-cryptanoside A, (-)-oleandrin, (-)-ouabain, and (+)-strebloside. It is hoped that this contribution will provide some helpful information for the design and discovery of new cardiac glycoside-type therapeutic agents for the treatment of cardiovascular diseases and cancer.
Collapse
Affiliation(s)
- Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| | - Andrew T Anderson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Gunnar Meyer
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Kaitlyn M Lauber
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Judith C Gallucci
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
14
|
El-Mahdy NA, Tadros MG, El-Masry TA, Binsaleh AY, Alsubaie N, Alrossies A, Abd Elhamid MI, Osman EY, Shalaby HM, Saif DS. Efficacy of the cardiac glycoside digoxin as an adjunct to csDMARDs in rheumatoid arthritis patients: a randomized, double-blind, placebo-controlled trial. Front Pharmacol 2024; 15:1445708. [PMID: 39498340 PMCID: PMC11532073 DOI: 10.3389/fphar.2024.1445708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/17/2024] [Indexed: 11/07/2024] Open
Abstract
Background Inflammation and angiogenesis are two main mechanisms that act as mutual pathways in rheumatoid arthritis (RA). This work aimed to study the efficacy of digoxin as an adjunct therapy to conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) in active RA patients. Methods In a randomized, double-blinded, placebo-controlled study, 60 adult patients with active RA received a placebo or digoxin (0.25 mg every other day) combined with csDMARDs for 6 months. The American College of Rheumatology (ACR) 20, ACR50, and ACR70 response rates and the disease activity score (DAS28) were assessed for patients. Flow cytometric analysis of Th17 cells and serum concentrations of IL-17A, IL-23, HIF-1α, and VEGF were evaluated before and after three and 6 months of therapy. Results Following three and 6 months of digoxin therapy combined with csDMARDs, significant differences were detected in laboratory and clinical parameters relative to the control group. After 6 months, 83.3% of patients in the digoxin group, compared to 56.7% in the control group, achieved an ACR20 response (p = 0.024). The digoxin group had a significantly higher percentage of patients who achieved DAS28 remission after 6 months (p = 0.024). Notable improvements in the Health Assessment Questionnaire Disability Index, ACR50, and ACR70 were detected in the digoxin group. Conclusion Digoxin was well tolerated and exerted profound immunomodulatory and anti-inflammatory effects in RA patients, and may also exhibit anti-angiogenic properties, indicating that it might be an effective adjunct to csDMARDs in treating RA. Clinical Trial Registration clinicaltrials.gov, identifier NCT04834557.
Collapse
Affiliation(s)
- Nageh A. El-Mahdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mariam G. Tadros
- Department of Pharmacy Practice, Faculty of Pharmacy, Sinai University - Arish Branch, Arish, Egypt
| | - Thanaa A. El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ammena Y. Binsaleh
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Nawal Alsubaie
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amani Alrossies
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Enas Y. Osman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Hadeel M. Shalaby
- Department of Clinical Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Dalia S. Saif
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| |
Collapse
|
15
|
Ha DP, Shin WJ, Liu Z, Doche ME, Lau R, Leli NM, Conn CS, Russo M, Lorenzato A, Koumenis C, Yu M, Mumenthaler SM, Lee AS. Targeting stress induction of GRP78 by cardiac glycoside oleandrin dually suppresses cancer and COVID-19. Cell Biosci 2024; 14:115. [PMID: 39238058 PMCID: PMC11378597 DOI: 10.1186/s13578-024-01297-3] [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: 04/16/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Despite recent therapeutic advances, combating cancer resistance remains a formidable challenge. The 78-kilodalton glucose-regulated protein (GRP78), a key stress-inducible endoplasmic reticulum (ER) chaperone, plays a crucial role in both cancer cell survival and stress adaptation. GRP78 is also upregulated during SARS-CoV-2 infection and acts as a critical host factor. Recently, we discovered cardiac glycosides (CGs) as novel suppressors of GRP78 stress induction through a high-throughput screen of clinically relevant compound libraries. This study aims to test the possibility that agents capable of blocking stress induction of GRP78 could dually suppress cancer and COVID-19. RESULTS Here we report that oleandrin (OLN), is the most potent among the CGs in inhibiting acute stress induction of total GRP78, which also results in reduced cell surface and nuclear forms of GRP78 in stressed cells. The inhibition of stress induction of GRP78 is at the post-transcriptional level, independent of protein degradation and autophagy and may involve translational control as OLN blocks stress-induced loading of ribosomes onto GRP78 mRNAs. Moreover, the human Na+/K+-ATPase α3 isoform is critical for OLN suppression of GRP78 stress induction. OLN, in nanomolar range, enhances apoptosis, sensitizes colorectal cancer cells to chemotherapeutic agents, and reduces the viability of patient-derived colon cancer organoids. Likewise, OLN, suppresses GRP78 expression and impedes tumor growth in an orthotopic breast cancer xenograft model. Furthermore, OLN blocks infection by SARS-CoV-2 and its variants and enhances existing anti-viral therapies. Notably, GRP78 overexpression mitigates OLN-mediated cancer cell apoptotic onset and suppression of virus release. CONCLUSION Our findings validate GRP78 as a target of OLN anti-cancer and anti-viral activities. These proof-of-principle studies support further investigation of OLN as a readily accessible compound to dually combat cancer and COVID-19.
Collapse
Affiliation(s)
- Dat P Ha
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Woo-Jin Shin
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, 34987, USA
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Ze Liu
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Michael E Doche
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Roy Lau
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Nektaria Maria Leli
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Crystal S Conn
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariangela Russo
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Annalisa Lorenzato
- Dipartimento di Oncologia, Molecular Biotechnology Center, Università di Torino, Turin, Italy
| | - Constantinos Koumenis
- Department of Radiation Oncology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Min Yu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shannon M Mumenthaler
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
16
|
Kumar PP, Smith D, Key J, Dong H, Ganapathysamy A, Maranda V, Wong NKY, Fernandez ML, Kim H, Zhang G, Ewanowich C, Hopkins L, Freywald A, Postovit LM, Köbel M, Fu Y, Vizeacoumar FS, Vizeacoumar FJ, Carey MS, Lee CH. Preclinical 3D model screening reveals digoxin as an effective therapy for a rare and aggressive type of endometrial cancer. Gynecol Oncol 2024; 188:162-168. [PMID: 38970843 DOI: 10.1016/j.ygyno.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVE Dedifferentiated endometrial carcinoma (DDEC) characterized by SWItch/Sucrose Non-Fermentable (SWI/SNF) complex inactivation is a highly aggressive type of endometrial cancer without effective systemic therapy options. Its uncommon nature and aggressive disease trajectory pose significant challenges for therapeutic progress. To address this obstacle, we focused on developing preclinical models tailored to this tumor type and established patient tumor-derived three-dimensional (3D) spheroid models of DDEC. METHODS High-throughput drug repurposing screens were performed on in vitro 3D spheroid models of DDEC cell lines (SMARCA4-inactivated DDEC-1 and ARID1A/ARID1B co-inactivated DDEC-2). The dose-response relationships of the identified candidate drugs were evaluated in vitro, followed by in vivo evaluation using xenograft models of DDEC-1 and DDEC-2. RESULTS Drug screen in 3D models identified multiple cardiac glycosides including digoxin and digitoxin as candidate drugs in both DDEC-1 and DDEC-2. Subsequent in vitro dose-response analyses confirmed the inhibitory activity of digoxin and digitoxin with both drugs showing lower IC50 in DDEC cells compared to non-DDEC endometrial cancer cells. In in vivo xenograft models, digoxin significantly suppressed the growth of DDEC tumors at clinically relevant serum concentrations. CONCLUSION Using biologically precise preclinical models of DDEC derived from patient tumor samples, our study identified digoxin as an effective drug in suppressing DDEC tumor growth. These findings provide compelling preclinical evidence for the use of digoxin as systemic therapy for SWI/SNF-inactivated DDEC, which may also be applicable to other SWI/SNF-inactivated tumor types.
Collapse
Affiliation(s)
- Pooja Praveen Kumar
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - DuPreez Smith
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - James Key
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - He Dong
- Division of Oncology, College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| | | | - Vincent Maranda
- Division of Oncology, College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| | - Nelson K Y Wong
- Department of Experimental Therapeutics, BC Cancer, Vancouver, British Columbia, Canada
| | | | - Hannah Kim
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - Guihua Zhang
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Carol Ewanowich
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Laura Hopkins
- Division of Oncology, Cancer Cluster, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, Canada
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Lynne M Postovit
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, Calgary Laboratory Services and University of Calgary, Calgary, Alberta, Canada
| | - Yangxin Fu
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Frederick S Vizeacoumar
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Franco J Vizeacoumar
- Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, Canada; Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Mark S Carey
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - Cheng-Han Lee
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
17
|
Huang Z, Tang Y, Zhang J, Huang J, Cheng R, Guo Y, Kleer CG, Wang Y, Xue L. Hypoxia makes EZH2 inhibitor not easy-advances of crosstalk between HIF and EZH2. LIFE METABOLISM 2024; 3:loae017. [PMID: 38911968 PMCID: PMC11192520 DOI: 10.1093/lifemeta/loae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/28/2024] [Accepted: 05/03/2024] [Indexed: 06/25/2024]
Abstract
Histone methylation plays a crucial role in tumorigenesis. Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that regulates chromatin structure and gene expression. EZH2 inhibitors (EZH2is) have been shown to be effective in treating hematologic malignancies, while their effectiveness in solid tumors remains limited. One of the major challenges in the treatment of solid tumors is their hypoxic tumor microenvironment. Hypoxia-inducible factor 1-alpha (HIF-1α) is a key hypoxia responder that interacts with EZH2 to promote tumor progression. Here we discuss the implications of the relationship between EZH2 and hypoxia for expanding the application of EZH2is in solid tumors.
Collapse
Affiliation(s)
- Zhanya Huang
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Yuanjun Tang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Jianlin Zhang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Jiaqi Huang
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Rui Cheng
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Yunyun Guo
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Yuqing Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Lixiang Xue
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
18
|
Qiu W, Zhao L, Liu H, Xu P, Qian C. Hypoxia-induced NOS1 as a therapeutic target in hypercholesterolemia-related colorectal cancer. Cancer Metab 2024; 12:14. [PMID: 38755702 PMCID: PMC11100240 DOI: 10.1186/s40170-024-00338-2] [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: 11/20/2023] [Accepted: 03/15/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND It is well established that hypercholesterolemia increases the risk of atherosclerosis, especially because it reduces the availability of nitric oxide (NO). However, the relationship between hypercholesterolemia and NO in regulating colorectal cancer development and progression remains unknown. METHODS We conducted bioinformatics analysis, qRT-PCR, ChIP-qPCR assays, luciferase report assays, clonogenic survival assays, and multiple mouse models to investigate the function and mechanism of hypercholesterolemia in regulating NO signaling. Additionally, NOS inhibitors were used to evaluate the potential of therapeutic strategy in anti-tumor response. RESULTS Here, we show that oxidized low-density lipoprotein (oxLDL) cholesterol and its receptor LOX-1 are essential for hypercholesterolemia-induced colorectal tumorigenesis. Mechanically, the oxLDL promotes the oxidant stress-dependent induction of hypoxia signaling to transcriptionally up-regulate NO synthase (NOS) especially NOS1 expression in colorectal cancer (CRC) cells. More importantly, our results suggested that selective inhibition of NOS1 with its specific inhibitor Nω-Propyl-L-arginine is a suitable therapeutic strategy for hypercholesterolemia-related CRC with both efficacy and toxicity reduction. CONCLUSIONS Our findings established that hypercholesterolemia induces the oxidant stress-dependent induction of hypoxia signaling to transcriptionally up-regulate NOS1 expression in CRC cells, and the clinically applicable NOS1 inhibitor Nω-Propyl-L-arginine represents an effective therapeutic strategy for hypercholesterolemia-related CRC.
Collapse
Affiliation(s)
- Weiqing Qiu
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2000 Jiangyue Road, Shanghai, 200012, China
| | - Li Zhao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, 200012, China
| | - Hua Liu
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2000 Jiangyue Road, Shanghai, 200012, China
| | - Ping Xu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, 200012, China.
| | - Changlin Qian
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2000 Jiangyue Road, Shanghai, 200012, China.
| |
Collapse
|
19
|
Moinuddin A, Poznanski SM, Portillo AL, Monteiro JK, Ashkar AA. Metabolic adaptations determine whether natural killer cells fail or thrive within the tumor microenvironment. Immunol Rev 2024; 323:19-39. [PMID: 38459782 DOI: 10.1111/imr.13316] [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] [Indexed: 03/10/2024]
Abstract
Natural Killer (NK) cells are a top contender in the development of adoptive cell therapies for cancer due to their diverse antitumor functions and ability to restrict their activation against nonmalignant cells. Despite their success in hematologic malignancies, NK cell-based therapies have been limited in the context of solid tumors. Tumor cells undergo various metabolic adaptations to sustain the immense energy demands that are needed to support their rapid and uncontrolled proliferation. As a result, the tumor microenvironment (TME) is depleted of nutrients needed to fuel immune cell activity and contains several immunosuppressive metabolites that hinder NK cell antitumor functions. Further, we now know that NK cell metabolic status is a main determining factor of their effector functions. Hence, the ability of NK cells to withstand and adapt to these metabolically hostile conditions is imperative for effective and sustained antitumor activity in the TME. With this in mind, we review the consequences of metabolic hostility in the TME on NK cell metabolism and function. We also discuss tumor-like metabolic programs in NK cell induced by STAT3-mediated expansion that adapt NK cells to thrive in the TME. Finally, we examine how other approaches can be applied to enhance NK cell metabolism in tumors.
Collapse
Affiliation(s)
- Adnan Moinuddin
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario, Canada
| | - Sophie M Poznanski
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario, Canada
| | - Ana L Portillo
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan K Monteiro
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario, Canada
| | - Ali A Ashkar
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
20
|
Murthy D, Attri KS, Suresh V, Rajacharya GH, Valenzuela CA, Thakur R, Zhao J, Shukla SK, Chaika NV, LaBreck D, Rao CV, Hollingsworth MA, Mehla K, Singh PK. The MUC1-HIF-1α signaling axis regulates pancreatic cancer pathogenesis through polyamine metabolism remodeling. Proc Natl Acad Sci U S A 2024; 121:e2315509121. [PMID: 38547055 PMCID: PMC10998584 DOI: 10.1073/pnas.2315509121] [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: 09/06/2023] [Accepted: 02/22/2024] [Indexed: 04/02/2024] Open
Abstract
Dysregulation of polyamine metabolism has been implicated in cancer initiation and progression; however, the mechanism of polyamine dysregulation in cancer is not fully understood. In this study, we investigated the role of MUC1, a mucin protein overexpressed in pancreatic cancer, in regulating polyamine metabolism. Utilizing pancreatic cancer patient data, we noted a positive correlation between MUC1 expression and the expression of key polyamine metabolism pathway genes. Functional studies revealed that knockdown of spermidine/spermine N1-acetyltransferase 1 (SAT1), a key enzyme involved in polyamine catabolism, attenuated the oncogenic functions of MUC1, including cell survival and proliferation. We further identified a regulatory axis whereby MUC1 stabilized hypoxia-inducible factor (HIF-1α), leading to increased SAT1 expression, which in turn induced carbon flux into the tricarboxylic acid cycle. MUC1-mediated stabilization of HIF-1α enhanced the promoter occupancy of the latter on SAT1 promoter and corresponding transcriptional activation of SAT1, which could be abrogated by pharmacological inhibition of HIF-1α or CRISPR/Cas9-mediated knockout of HIF1A. MUC1 knockdown caused a significant reduction in the levels of SAT1-generated metabolites, N1-acetylspermidine and N8-acetylspermidine. Given the known role of MUC1 in therapy resistance, we also investigated whether inhibiting SAT1 would enhance the efficacy of FOLFIRINOX chemotherapy. By utilizing organoid and orthotopic pancreatic cancer mouse models, we observed that targeting SAT1 with pentamidine improved the efficacy of FOLFIRINOX, suggesting that the combination may represent a promising therapeutic strategy against pancreatic cancer. This study provides insights into the interplay between MUC1 and polyamine metabolism, offering potential avenues for the development of treatments against pancreatic cancer.
Collapse
Affiliation(s)
- Divya Murthy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Kuldeep S. Attri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Voddu Suresh
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Girish H. Rajacharya
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Carlos A. Valenzuela
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Ravi Thakur
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Junzhang Zhao
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Surendra K. Shukla
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Nina V. Chaika
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Drew LaBreck
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chinthalapally V. Rao
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Kamiya Mehla
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Pankaj K. Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| |
Collapse
|
21
|
Pandey S, Singh R, Habib N, Tripathi RM, Kushwaha R, Mahdi AA. Regulation of Hypoxia Dependent Reprogramming of Cancer Metabolism: Role of HIF-1 and Its Potential Therapeutic Implications in Leukemia. Asian Pac J Cancer Prev 2024; 25:1121-1134. [PMID: 38679971 PMCID: PMC11162727 DOI: 10.31557/apjcp.2024.25.4.1121] [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: 08/18/2023] [Accepted: 04/20/2024] [Indexed: 05/01/2024] Open
Abstract
Metabolic reprogramming occurs to meet cancer cells' high energy demand. Its function is essential to the survival of malignancies. Comparing cancer cells to non-malignant cells has revealed that cancer cells have altered metabolism. Several pathways, particularly mTOR, Akt, PI3K, and HIF-1 (hypoxia-inducible factor-1) modulate the metabolism of cancer. Among other aspects of cancer biology, gene expression in metabolism, survival, invasion, proliferation, and angiogenesis of cells are controlled by HIF-1, a vital controller of cellular responsiveness to hypoxia. This article examines various cancer cell metabolisms, metabolic alterations that can take place in cancer cells, metabolic pathways, and molecular aspects of metabolic alteration in cancer cells placing special attention on the consequences of hypoxia-inducible factor and summarising some of their novel targets in the treatment of cancer including leukemia. A brief description of HIF-1α's role and target in a few common types of hematological malignancies (leukemia) is also elucidated in the present article.
Collapse
Affiliation(s)
- Sandeep Pandey
- Department of Biochemistry, King George’s Medical University, Lucknow, U.P., India.
| | - Ranjana Singh
- Department of Biochemistry, King George’s Medical University, Lucknow, U.P., India.
| | - Nimra Habib
- Department of Biochemistry, King George’s Medical University, Lucknow, U.P., India.
| | - Ramesh Mani Tripathi
- Department of Biochemistry, King George’s Medical University, Lucknow, U.P., India.
| | - Rashmi Kushwaha
- Department of Pathology, King George’s Medical University, Lucknow, U.P., India.
| | - Abbas Ali Mahdi
- Department of Biochemistry, King George’s Medical University, Lucknow, U.P., India.
| |
Collapse
|
22
|
Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev 2024; 43:501-574. [PMID: 37792223 DOI: 10.1007/s10555-023-10136-9] [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: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Tumor cells employ multiple signaling mediators to escape the hypoxic condition and trigger angiogenesis and metastasis. As a critical orchestrate of tumorigenic conditions, hypoxia-inducible factor-1 (HIF-1) is responsible for stimulating several target genes and dysregulated pathways in tumor invasion and migration. Therefore, targeting HIF-1 pathway and cross-talked mediators seems to be a novel strategy in cancer prevention and treatment. In recent decades, tremendous efforts have been made to develop multi-targeted therapies to modulate several dysregulated pathways in cancer angiogenesis, invasion, and metastasis. In this line, natural compounds have shown a bright future in combating angiogenic and metastatic conditions. Among the natural secondary metabolites, we have evaluated the critical potential of phenolic compounds, terpenes/terpenoids, alkaloids, sulfur compounds, marine- and microbe-derived agents in the attenuation of HIF-1, and interconnected pathways in fighting tumor-associated angiogenesis and invasion. This is the first comprehensive review on natural constituents as potential regulators of HIF-1 and interconnected pathways against cancer angiogenesis and metastasis. This review aims to reshape the previous strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6714415153, Iran
| | - Kassidy Webber
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA.
| |
Collapse
|
23
|
Yuan X, Ruan W, Bobrow B, Carmeliet P, Eltzschig HK. Targeting hypoxia-inducible factors: therapeutic opportunities and challenges. Nat Rev Drug Discov 2024; 23:175-200. [PMID: 38123660 DOI: 10.1038/s41573-023-00848-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Hypoxia-inducible factors (HIFs) are highly conserved transcription factors that are crucial for adaptation of metazoans to limited oxygen availability. Recently, HIF activation and inhibition have emerged as therapeutic targets in various human diseases. Pharmacologically desirable effects of HIF activation include erythropoiesis stimulation, cellular metabolism optimization during hypoxia and adaptive responses during ischaemia and inflammation. By contrast, HIF inhibition has been explored as a therapy for various cancers, retinal neovascularization and pulmonary hypertension. This Review discusses the biochemical mechanisms that control HIF stabilization and the molecular strategies that can be exploited pharmacologically to activate or inhibit HIFs. In addition, we examine medical conditions that benefit from targeting HIFs, the potential side effects of HIF activation or inhibition and future challenges in this field.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Wei Ruan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Anaesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bentley Bobrow
- Department of Emergency Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Holger K Eltzschig
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Outcomes Research Consortium, Cleveland, OH, USA.
| |
Collapse
|
24
|
Bayona C, Alza L, Ranđelović T, Sallán MC, Visa A, Cantí C, Ochoa I, Oliván S, Herreros J. Tetralol derivative NNC-55-0396 targets hypoxic cells in the glioblastoma microenvironment: an organ-on-chip approach. Cell Death Dis 2024; 15:127. [PMID: 38341408 PMCID: PMC10858941 DOI: 10.1038/s41419-024-06492-1] [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: 07/27/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
Glioblastoma (GBM) is a highly malignant brain tumour characterised by limited treatment options and poor prognosis. The tumour microenvironment, particularly the central hypoxic region of the tumour, is known to play a pivotal role in GBM progression. Cells within this region adapt to hypoxia by stabilising transcription factor HIF1-α, which promotes cell proliferation, dedifferentiation and chemoresistance. In this study we sought to examine the effects of NNC-55-0396, a tetralol compound which overactivates the unfolded protein response inducing apoptosis, using the organ-on-chip technology. We identified an increased sensitivity of the hypoxic core of the chip to NNC, which correlates with decreasing levels of HIF1-α in vitro. Moreover, NNC blocks the macroautophagic process that is unleashed by hypoxia as revealed by increased levels of autophagosomal constituent LC3-II and autophagy chaperone p62/SQSTM1. The specific effects of NNC in the hypoxic microenvironment unveil additional anti-cancer abilities of this compound and further support investigations on its use in combined therapies against GBM.
Collapse
Affiliation(s)
- Clara Bayona
- Tissue Microenvironment (TME) Lab, Institute for Health Research Aragón (IIS Aragón), Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018, Zaragoza, Spain
| | - Lía Alza
- Calcium Cell Signaling, IRBLleida, University of Lleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Teodora Ranđelović
- Tissue Microenvironment (TME) Lab, Institute for Health Research Aragón (IIS Aragón), Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 50018, Zaragoza, Spain
| | - Marta C Sallán
- Calcium Cell Signaling, IRBLleida, University of Lleida, Rovira Roure 80, 25198, Lleida, Spain
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Anna Visa
- Calcium Cell Signaling, IRBLleida, University of Lleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Carles Cantí
- Calcium Cell Signaling, IRBLleida, University of Lleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Institute for Health Research Aragón (IIS Aragón), Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 50018, Zaragoza, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Institute for Health Research Aragón (IIS Aragón), Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018, Zaragoza, Spain.
| | - Judit Herreros
- Calcium Cell Signaling, IRBLleida, University of Lleida, Rovira Roure 80, 25198, Lleida, Spain.
| |
Collapse
|
25
|
Bokhari SZ, Aloss K, Leroy Viana PH, Schvarcz CA, Besztercei B, Giunashvili N, Bócsi D, Koós Z, Balogh A, Benyó Z, Hamar P. Digoxin-Mediated Inhibition of Potential Hypoxia-Related Angiogenic Repair in Modulated Electro-Hyperthermia (mEHT)-Treated Murine Triple-Negative Breast Cancer Model. ACS Pharmacol Transl Sci 2024; 7:456-466. [PMID: 38357275 PMCID: PMC10863435 DOI: 10.1021/acsptsci.3c00296] [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: 10/23/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 02/16/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer type with no targeted therapy and hence limited treatment options. Modulated electrohyperthermia (mEHT) is a novel complementary therapy where a 13.56 MHz radiofrequency current targets cancer cells selectively, inducing tumor damage by thermal and electromagnetic effects. We observed severe vascular damage in mEHT-treated tumors and investigated the potential synergism between mEHT and inhibition of tumor vasculature recovery in our TNBC mouse model. 4T1/4T07 isografts were orthotopically inoculated and treated three to five times with mEHT. mEHT induced vascular damage 4-12 h after treatment, leading to tissue hypoxia detected at 24 h. Hypoxia in treated tumors induced an angiogenic recovery 24 h after the last treatment. Administration of the cardiac glycoside digoxin with the potential hypoxia-inducible factor 1-α (HIF1-α) and angiogenesis inhibitory effects could synergistically augment mEHT-mediated tumor damage and reduce tissue hypoxia signaling and consequent vascular recovery in mEHT-treated TNBC tumors. Conclusively, repeated mEHT induced vascular damage and hypoxic stress in TNBC that promoted vascular recovery. Inhibiting this hypoxic stress signaling enhanced the effectiveness of mEHT and may potentially enhance other forms of cancer treatment.
Collapse
Affiliation(s)
| | - Kenan Aloss
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | | | - Csaba András Schvarcz
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
- Cerebrovascular
and Neurocognitive Disorders Research Group, Eötvös, Loránd Research Network and Semmelweis
University (ELKH-SE), Tűzoltó utca 37-47, Budapest 1094, Hungary
| | - Balázs Besztercei
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Nino Giunashvili
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Dániel Bócsi
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Zoltán Koós
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Andrea Balogh
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Zoltán Benyó
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Péter Hamar
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| |
Collapse
|
26
|
Ortmann BM. Hypoxia-inducible factor in cancer: from pathway regulation to therapeutic opportunity. BMJ ONCOLOGY 2024; 3:e000154. [PMID: 39886164 PMCID: PMC11203102 DOI: 10.1136/bmjonc-2023-000154] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2025]
Abstract
Cancer remains one of the most formidable challenges in modern medicine, due to its complex and dynamic nature, which demands innovative therapeutic approaches. One major challenge to cancer treatment is the tumour microenvironment and in particular tumour hypoxia (low oxygen levels), which contributes to tumour progression and immune evasion. At the cellular level, this is primarily governed by hypoxia-inducible factor (HIF). HIF is a transcription factor that orchestrates cellular responses to low oxygen levels, driving angiogenesis, metabolic adaptation and immune regulation. HIF's dysregulation is frequently observed in various cancer types and correlates with increased aggressiveness, metastasis, resistance to therapy and poor patient prognosis. Consequently, understanding the cellular mechanisms underlying HIF activation and its downstream effects has become crucial to developing targeted cancer therapies for improving cancer patient outcomes and represents a key step towards precision medicine. Recent advancements in drug development have led to the emergence of HIF inhibitors, which aim to disrupt HIF-driven processes in cancer providing therapeutic benefit. Here, we provide a review of the molecular mechanisms through which HIF promotes tumour growth and resistance, emphasising the potential clinical benefits of HIF-targeted therapies. This review will discuss the challenges and opportunities associated with translating HIF inhibition into clinical practice, including ongoing clinical trials and future directions in the development of HIF-based cancer treatments.
Collapse
Affiliation(s)
- Brian M Ortmann
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
27
|
Jin Y, Peng Y, Xu J, Yuan Y, Yang N, Zhang Z, Xu L, Li L, Xiong Y, Sun D, Pan Y, Wu R, Fu J. LUBAC promotes angiogenesis and lung tumorigenesis by ubiquitinating and antagonizing autophagic degradation of HIF1α. Oncogenesis 2024; 13:6. [PMID: 38272870 PMCID: PMC10810860 DOI: 10.1038/s41389-024-00508-3] [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: 09/30/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Hypoxia-inducible factor 1 (HIF1) is critically important for driving angiogenesis and tumorigenesis. Linear ubiquitin chain assembly complex (LUBAC), the only known ubiquitin ligase capable of catalyzing protein linear ubiquitination to date, is implicated in cell signaling and associated with cancers. However, the role and mechanism of LUBAC in regulating the expression and function of HIF1α, the labile subunit of HIF1, remain to be elucidated. Herein we showed that LUBAC increases HIF1α protein expression in cultured cells and tissues of human lung cancer and enhances HIF1α DNA-binding and transcriptional activities, which are dependent upon LUBAC enzymatic activity. Mechanistically, LUBAC increases HIF1α stability through antagonizing HIF1α decay by the chaperone-mediated autophagy (CMA)-lysosome pathway, thereby potentiating HIF1α activity. We further demonstrated that HIF1α selectively interacts with HOIP (the catalytic subunit of LUBAC) primarily in the cytoplasm. LUBAC catalyzes linear ubiquitination of HIF1α at lysine 362. Linear ubiquitination shields HIF1α from interacting with heat-shock cognate protein of 70 kDa and lysosome-associated membrane protein type 2 A, two components of CMA. Consequently, linear ubiquitination confers protection against CMA-mediated destruction of HIF1α, increasing HIF1α stability and activity. We found that prolyl hydroxylation is not a perquisite for LUBAC's effects on HIF1α. Functionally, LUBAC facilitates proliferation, clonogenic formation, invasion and migration of lung cancer cells. LUBAC also boosts angiogenesis and exacerbates lung cancer growth in mice, which are greatly compromised by inhibition of HIF1α. This work provides novel mechanistic insights into the role of LUBAC in regulating HIF1α homeostasis, tumor angiogenesis and tumorigenesis of lung cancer, making LUBAC an attractive therapeutic target for cancers.
Collapse
Affiliation(s)
- Ying Jin
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China.
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Yazhi Peng
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Jie Xu
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Ye Yuan
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Yang
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Zemei Zhang
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
- Graduate School, Jinzhou Medical University, Liaoning, China
| | - Lei Xu
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Lin Li
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Yulian Xiong
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Dejiao Sun
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Yamu Pan
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Ruiqing Wu
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China
| | - Jian Fu
- The Laboratory of Inflammation and Vascular Biology, Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei, China.
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
28
|
Liu J, Jiang Y, Chen L, Qian Z, Zhang Y. Associations between HIFs and tumor immune checkpoints: mechanism and therapy. Discov Oncol 2024; 15:2. [PMID: 38165484 PMCID: PMC10761656 DOI: 10.1007/s12672-023-00836-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Hypoxia, which activates a variety of signaling pathways to enhance tumor cell growth and metabolism, is among the primary features of tumor cells. Hypoxia-inducible factors (HIFs) have a substantial impact on a variety of facets of tumor biology, such as epithelial-mesenchymal transition, metabolic reprogramming, angiogenesis, and improved radiation resistance. HIFs induce hypoxia-adaptive responses in tumor cells. Many academics have presented preclinical and clinical research targeting HIFs in tumor therapy, highlighting the potential applicability of targeted HIFs. In recent years, the discovery of numerous pharmacological drugs targeting the regulatory mechanisms of HIFs has garnered substantial attention. Additionally, HIF inhibitors have attained positive results when used in conjunction with traditional oncology radiation and/or chemotherapy, as well as with the very promising addition of tumor immunotherapy. Immune checkpoint inhibitors (CPIs), which are employed in a range of cancer treatments over the past decades, are essential in tumor immunotherapy. Nevertheless, the use of immunotherapy has been severely hampered by tumor resistance and treatment-related toxicity. According to research, HIF inhibitors paired with CPIs may be game changers for multiple malignancies, decreasing malignant cell plasticity and cancer therapy resistance, among other things, and opening up substantial new pathways for immunotherapy drug development. The structure, activation mechanisms, and pharmacological sites of action of the HIF family are briefly reviewed in this work. This review further explores the interactions between HIF inhibitors and other tumor immunotherapy components and covers the potential clinical use of HIF inhibitors in combination with CPIs.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Ying Jiang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Lingyan Chen
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Zhiwen Qian
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China.
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, 214000, Jiangsu, China.
| |
Collapse
|
29
|
Babapoor-Farrokhran S, Qin Y, Flores-Bellver M, Niu Y, Bhutto IA, Aparicio-Domingo S, Guo C, Rodrigues M, Domashevich T, Deshpande M, Megarity H, Chopde R, Eberhart CG, Canto-Soler V, Montaner S, Sodhi A. Pathologic vs. protective roles of hypoxia-inducible factor 1 in RPE and photoreceptors in wet vs. dry age-related macular degeneration. Proc Natl Acad Sci U S A 2023; 120:e2302845120. [PMID: 38055741 PMCID: PMC10723156 DOI: 10.1073/pnas.2302845120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 09/20/2023] [Indexed: 12/08/2023] Open
Abstract
It has previously been reported that antioxidant vitamins can help reduce the risk of vision loss associated with progression to advanced age-related macular degeneration (AMD), a leading cause of visual impairment among the elderly. Nonetheless, how oxidative stress contributes to the development of choroidal neovascularization (CNV) in some AMD patients and geographic atrophy (GA) in others is poorly understood. Here, we provide evidence demonstrating that oxidative stress cooperates with hypoxia to synergistically stimulate the accumulation of hypoxia-inducible factor (HIF)-1α in the retinal pigment epithelium (RPE), resulting in increased expression of the HIF-1-dependent angiogenic mediators that promote CNV. HIF-1 inhibition blocked the expression of these angiogenic mediators and prevented CNV development in an animal model of ocular oxidative stress, demonstrating the pathological role of HIF-1 in response to oxidative stress stimulation in neovascular AMD. While human-induced pluripotent stem cell (hiPSC)-derived RPE monolayers exposed to chemical oxidants resulted in disorganization and disruption of their normal architecture, RPE cells proved remarkably resistant to oxidative stress. Conversely, equivalent doses of chemical oxidants resulted in apoptosis of hiPSC-derived retinal photoreceptors. Pharmacologic inhibition of HIF-1 in the mouse retina enhanced-while HIF-1 augmentation reduced-photoreceptor apoptosis in two mouse models for oxidative stress, consistent with a protective role for HIF-1 in photoreceptors in patients with advanced dry AMD. Collectively, these results suggest that in patients with AMD, increased expression of HIF-1α in RPE exposed to oxidative stress promotes the development of CNV, but inadequate HIF-1α expression in photoreceptors contributes to the development of GA.
Collapse
Affiliation(s)
| | - Yu Qin
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang110005, China
- Department of Ophthalmology, Eye Hospital of China Medical University, Shenyang110005, China
- Key Lens Research Laboratory of Liaoning Province, Shenyang110005, China
| | - Miguel Flores-Bellver
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO80045
| | - Yueqi Niu
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Imran A. Bhutto
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Silvia Aparicio-Domingo
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO80045
| | - Chuanyu Guo
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Murilo Rodrigues
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Timothy Domashevich
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO80045
| | - Monika Deshpande
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Haley Megarity
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Rakesh Chopde
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Charles G. Eberhart
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO80045
| | - Silvia Montaner
- Department of Oncology and Diagnostic Sciences, Greenebaum Cancer Center, University of Maryland, Baltimore, MD21201
| | - Akrit Sodhi
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD21287
| |
Collapse
|
30
|
Xiang F, Luo F. Stem cell factor modulates HIF-1α levels and diminishes 5-FU sensitivity in 5-FU resistant pancreatic cells by altering the anabolic glucose metabolism. J Biochem Mol Toxicol 2023; 37:e23487. [PMID: 37718545 DOI: 10.1002/jbt.23487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/19/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023]
Abstract
Resistance to chemotherapy in cancer leads to poor therapeutic outcomes and also leads to challenges in treatment. The present work evaluated the mechanism involved in the resistance of 5-flurouracil (5-FU) in pancreatic cancer. At least 14 different pancreatic cancer (PC) cell lines were used for the study. For in vivo study female nude mice were selected. Patient-derived tumor xenograft samples were obtained from patients. The study involved, study for glucose uptake, fluorescence-activated cell sorting for glucose transporter, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide for cell survival, Picto-micrography for clonogenic assay, glutamine uptake assay, extracellular acidification and oxygen consumption rate, carbon dioxide release assay and lactate assay were also done. In addition to this, quantitative real-time polymerase chain reaction analysis for expression of genes, chromatin immunoprecipitation assay, western blot for protein expression, and immunohistochemical analysis in tumor sections, the tumors were studied by imaging for hypoxia and localization of TKT and CTPS-2. Also, patient-derived xenograft tumors were engrafted in nude mice, followed by a glucose uptake assay. We reported that elevated glycolytic flux causes dependence on glucose in cancer cells and, at the same time, increases pyrimidine biosynthesis. It was also found that stem cell factor-mediated stabilization of hypoxia-inducible factor-1a (HIF-1α) modulates the resistance in PC. Targeting HIF-1α in combination with 5-FU, strongly reduced the tumor burden. The study concludes that stem cell factor modulates HIF-1α and decreases the sensitivity in 5-FU resistant pancreatic cancer cells by targeting glucose metabolism. Deceased expression levels of CTPS-2 and TKT, which are regulators of pyrimidine biosynthesis could better the chance of survival in patients of pancreatic cancer receiving treatment of 5-FU.
Collapse
Affiliation(s)
- Fu Xiang
- Department of General Surgery, Dalian Medical University, Dalian, Liaoning, China
| | - Fuwen Luo
- Department of Acute Abdominal Surgery, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
31
|
Kang GS, Jo HJ, Lee YR, Oh T, Park HJ, Ahn GO. Sensing the oxygen and temperature in the adipose tissues - who's sensing what? Exp Mol Med 2023; 55:2300-2307. [PMID: 37907745 PMCID: PMC10689767 DOI: 10.1038/s12276-023-01113-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 11/02/2023] Open
Abstract
Adipose tissues, composed of various cell types, including adipocytes, endothelial cells, neurons, and immune cells, are organs that are exposed to dynamic environmental challenges. During diet-induced obesity, white adipose tissues experience hypoxia due to adipocyte hypertrophy and dysfunctional vasculature. Under these conditions, cells in white adipose tissues activate hypoxia-inducible factor (HIF), a transcription factor that activates signaling pathways involved in metabolism, angiogenesis, and survival/apoptosis to adapt to such an environment. Exposure to cold or activation of the β-adrenergic receptor (through catecholamines or chemicals) leads to heat generation, mainly in brown adipose tissues through activating uncoupling protein 1 (UCP1), a proton uncoupler in the inner membrane of the mitochondria. White adipose tissues can undergo a similar process under this condition, a phenomenon known as 'browning' of white adipose tissues or 'beige adipocytes'. While UCP1 expression has largely been confined to adipocytes, HIF can be expressed in many types of cells. To dissect the role of HIF in specific types of cells during diet-induced obesity, researchers have generated tissue-specific knockout (KO) mice targeting HIF pathways, and many studies have commonly revealed that intact HIF-1 signaling in adipocytes and adipose tissue macrophages exacerbates tissue inflammation and insulin resistance. In this review, we highlight some of the key findings obtained from these transgenic mice, including Ucp1 KO mice and other models targeting the HIF pathway in adipocytes, macrophages, or endothelial cells, to decipher their roles in diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Ye-Rim Lee
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Taerim Oh
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Joon Park
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
| |
Collapse
|
32
|
Isaacs JT, Dalrymple SL, Antony L, Rosen DM, Coleman IM, Nelson PS, Kostova M, Murray IA, Perdew GH, Denmeade SR, Akinboye ES, Brennen WN. Third generation quinoline-3-carboxamide transcriptional disrupter of HDAC4, HIF-1α, and MEF-2 signaling for metastatic castration-resistant prostate cancer. Prostate 2023; 83:1470-1493. [PMID: 37559436 PMCID: PMC10559933 DOI: 10.1002/pros.24606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/15/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND The quinoline-3-carboxamide, Tasquinimod (TasQ), is orally active as a maintenance therapy with an on-target mechanism-of-action via allosteric binding to HDAC4. This prevents formation of the HDAC4/NCoR1/HDAC3 complex, disrupting HIF-1α transcriptional activation and repressing MEF-2 target genes needed for adaptive survival signaling in the compromised tumor micro environment. In phase 3 clinical testing against metastatic castration-resistant prostate cancer(mCRPC), TasQ (1 mg/day) increased time-to-progression, but not overall survival. METHODS TasQ analogs were chemically synthesized and tested for activity compared to the parental compound. These included HDAC4 enzymatic assays, qRT-PCR and western blot analyses of gene and protein expression following treatment, in vitro and in vivo efficacy against multiple prostate cancer models including PDXs, pharmacokinetic analyses,AHR binding and agonist assays, SPR analyses of binding to HDAC4 and NCoR1, RNAseq analysis of in vivo tumors, 3D endothelial sprouting assays, and a targeted kinase screen. Genetic knockout or knockdown controls were used when appropriate. RESULTS Here, we document that, on this regimen (1 mg/day), TasQ blood levels are 10-fold lower than the optimal concentration (≥2 μM) needed for anticancer activity, suggesting higher daily doses are needed. Unfortunately, we also demonstrate that TasQ is an arylhydrocarbon receptor (AHR) agonist, which binds with an EC50 of 1 μM to produce unwanted off-target side effects. Therefore, we screened a library of TasQ analogsto maximize on-target versus off-target activity. Using this approach, we identified ESATA-20, which has ~10-fold lower AHR agonism and 5-fold greater potency against prostate cancer patient-derived xenografts. CONCLUSION This increased therapeuticindex nominates ESATA-20 as a lead candidate forclinical development as an orally active third generation quinoline-3-carboxamide analog thatretains its on-target ability to disrupt HDAC4/HIF-1α/MEF-2-dependent adaptive survival signaling in the compromisedtumor microenvironment found in mCRPC.
Collapse
Affiliation(s)
- John T. Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susan L. Dalrymple
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
| | - Lizamma Antony
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
| | - D. Marc Rosen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
| | - Ilsa M. Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Maya Kostova
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
| | - Iain A. Murray
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA
| | - Gary H. Perdew
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA
| | - Samuel R. Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Emmanuel S. Akinboye
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
| | - W. Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Carlos ACAM, Lemos JVM, Borges MMF, Albuquerque MCP, Sousa FB, Alves APNN, Dantas TS, Silva PGDB. Interleukin-17 plays a role in dental pulp inflammation mediated by zoledronic acid: a mechanism unrelated to the Th17 immune response? J Appl Oral Sci 2023; 31:e20230230. [PMID: 37820184 PMCID: PMC10567106 DOI: 10.1590/1678-7757-2023-0230] [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: 07/04/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVE To evaluate the influence of RORγT inhibition by digoxin on inflammatory changes related to interleukin-17 (IL-17) in the pulp of rats treated with zoledronate (ZOL). METHODOLOGY Forty male Wistar rats were divided into a negative control group (NCG) treated with saline solution, a positive control group (PCG) treated with ZOL (0.20 mg/kg), and three groups treated with ZOL and co-treated with digoxin 1, 2, or 4 mg/kg (DG1, 2, and 4). After four intravenous administrations of ZOL or saline solution in a 70-day protocol, the right molars were evaluated by histomorphometry (number of blood vessels, blood vessels/µm2, cells/µm2, total blood vessel area, and average blood vessel area) and immunohistochemistry (IL-17, TNF-α, IL-6, and TGF-β). The Kruskal-Wallis/Dunn test was used for statistical analysis. RESULTS PCG showed an increase in total blood vessel area (p=0.008) and average blood vessel area (p=0.014), and digoxin treatment reversed these changes. DG4 showed a reduction in blood vessels/µm2 (p<0.001). In PCG odontoblasts, there was an increase in IL-17 (p=0.002) and TNF-α (p=0.002) immunostaining, and in DG4, these changes were reversed. Odontoblasts in the digoxin-treated groups also showed an increase in IL-6 immunostaining (p<0.001) and a reduction in TGF-β immunostaining (p=0.002), and all ZOL-treated groups showed an increase in IL-17 (p=0.011) and TNF-α (p=0.017) in non-odontoblasts cells. CONCLUSION ZOL induces TNF-α- and IL-17-dependent vasodilation and ectasia, and the classical Th17 response activation pathway does not seem to participate in this process.
Collapse
Affiliation(s)
| | - José Vitor Mota Lemos
- Universidade Federal do Ceará, Departmento de Patologia Oral, Fortaleza, Ceará, Brasil
| | | | | | - Fabrício Bitu Sousa
- Universidade Federal do Ceará, Departmento de Patologia Oral, Fortaleza, Ceará, Brasil
- Centro Universitário Christus, Departamento de Patologia, Fortaleza, Ceará, Brasil
| | | | - Thinali Sousa Dantas
- Centro Universitário Christus, Departamento de Patologia, Fortaleza, Ceará, Brasil
| | - Paulo Goberlânio de Barros Silva
- Universidade Federal do Ceará, Departmento de Patologia Oral, Fortaleza, Ceará, Brasil
- Centro Universitário Christus, Departamento de Patologia, Fortaleza, Ceará, Brasil
| |
Collapse
|
34
|
Agamia NF, Sorror OA, Sayed NM, Ghazala RA, Echy SM, Moussa DH, Melnik BC. Overexpression of hypoxia-inducible factor-1α in hidradenitis suppurativa: the link between deviated immunity and metabolism. Arch Dermatol Res 2023; 315:2107-2118. [PMID: 36961533 PMCID: PMC10366312 DOI: 10.1007/s00403-023-02594-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/13/2023] [Accepted: 02/23/2023] [Indexed: 03/25/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is the master transcription factor of glycolysis, Th17 cell differentiation and suppression of regulatory T cells. In the skin and serum of patients with psoriasis vulgaris, increased expression of HIF-1α has been reported, whereas HIF-1α expression in the skin and serum of patients with hidradenitis suppurativa (HS) has not yet been studied. The objective of the study is to demonstrate is there a role for HIF-1α in the pathogenesis of hidradenitis suppurativa, and its relation to HS severity. Twenty patients suffering from hidradenitis suppurativa were included in the study. Punch biopsies were taken from lesional skin for the determination of HIF-1α expression by immunohistochemical staining, and HIF-1α gene expression by quantitative reverse transcription real time PCR. Quantification of HIF-1α protein concentration was done by enzyme-linked immunosorbent assay. Twenty socio-demographically cross-matched healthy volunteers served as controls. We found increased serum levels of HIF-1α. Literature-derived evidence indicates that the major clinical triggering factors of HS, obesity, and smoking are associated with hypoxia and enhanced HIF-1α expression. Pro-inflammatory cytokines such as tumor necrosis factor-[Formula: see text] via upregulation of nuclear factor [Formula: see text]B enhance HIF-1α expression. HIF-1α plays an important role for keratinocyte proliferation, especially for keratinocytes of the anagen hair follicle, which requires abundant glycolysis providing sufficient precursors molecules for biosynthetic pathways. Metformin via inhibition of mTORC1 as well as adalimumab attenuate HIF-1α expression, the key mediator between Th17-driven deviated immunity and keratinocyte hyperproliferation. In accordance with psoriasis, our study identifies HS as an HIF-1α-driven inflammatory skin disease and offers a new rationale for the prevention and treatment of HS by targeting HIF-1[Formula: see text] overexpression.
Collapse
Affiliation(s)
- Naglaa Fathi Agamia
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt.
| | - Osama Ahmed Sorror
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Naglaa Mohamed Sayed
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Rasha Abdelmawla Ghazala
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Sammar Mohamed Echy
- Department of Clinical Pathology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Doaa Helmy Moussa
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Bodo Clemens Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
35
|
Davis DA, Shrestha P, Yarchoan R. Hypoxia and hypoxia-inducible factors in Kaposi sarcoma-associated herpesvirus infection and disease pathogenesis. J Med Virol 2023; 95:e29071. [PMID: 37665216 PMCID: PMC10502919 DOI: 10.1002/jmv.29071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi sarcoma and several other tumors and hyperproliferative diseases seen predominantly in human immunodeficiency virus-infected and other immunocompromised persons. There is an increasing body of evidence showing that hypoxia and hypoxia-inducible factors (HIFs) play important roles in the biology of KSHV and in the pathogenesis of KSHV-induced diseases. Hypoxia and HIFs can induce lytic activation of KSHV and KSHV can in turn lead to a hypoxic-like state in infected cells. In this review, we describe the complex interactions between KSHV biology, the cellular responses to hypoxia, and the pathogenesis of KSHV-induced diseases. We also describe how interference with HIFs can lead to decreased tumor growth and/or death of infected cells and KSHV-induced tumors. Finally, we show how these observations may lead to novel strategies for the treatment of KSHV-induced diseases.
Collapse
Affiliation(s)
- David A Davis
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Prabha Shrestha
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Sakai N, Kamimura K, Terai S. Repurposable Drugs for Immunotherapy and Strategies to Find Candidate Drugs. Pharmaceutics 2023; 15:2190. [PMID: 37765160 PMCID: PMC10536625 DOI: 10.3390/pharmaceutics15092190] [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: 07/03/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Conventional drug discovery involves significant steps, time, and expenses; therefore, novel methods for drug discovery remain unmet, particularly for patients with intractable diseases. For this purpose, the drug repurposing method has been recently used to search for new therapeutic agents. Repurposed drugs are mostly previously approved drugs, which were carefully tested for their efficacy for other diseases and had their safety for the human body confirmed following careful pre-clinical trials, clinical trials, and post-marketing surveillance. Therefore, using these approved drugs for other diseases that cannot be treated using conventional therapeutic methods could save time and economic costs for testing their clinical applicability. In this review, we have summarized the methods for identifying repurposable drugs focusing on immunotherapy.
Collapse
Affiliation(s)
- Norihiro Sakai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan; (N.S.); (S.T.)
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan; (N.S.); (S.T.)
- Department of General Medicine, Niigata University School of Medicine, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan; (N.S.); (S.T.)
| |
Collapse
|
37
|
Zhang J, Sharma D, Dinabandhu A, Sanchez J, Applewhite B, Jee K, Deshpande M, Flores-Bellver M, Hu MW, Guo C, Salman S, Hwang Y, Anders NM, Rudek MA, Qian J, Canto-Soler MV, Semenza GL, Montaner S, Sodhi A. Targeting hypoxia-inducible factors with 32-134D safely and effectively treats diabetic eye disease in mice. J Clin Invest 2023; 133:e163290. [PMID: 37227777 PMCID: PMC10313368 DOI: 10.1172/jci163290] [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: 07/12/2022] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Many patients with diabetic eye disease respond inadequately to anti-VEGF therapies, implicating additional vasoactive mediators in its pathogenesis. We demonstrate that levels of angiogenic proteins regulated by HIF-1 and -2 remain elevated in the eyes of people with diabetes despite treatment with anti-VEGF therapy. Conversely, by inhibiting HIFs, we normalized the expression of multiple vasoactive mediators in mouse models of diabetic eye disease. Accumulation of HIFs and HIF-regulated vasoactive mediators in hyperglycemic animals was observed in the absence of tissue hypoxia, suggesting that targeting HIFs may be an effective early treatment for diabetic retinopathy. However, while the HIF inhibitor acriflavine prevented retinal vascular hyperpermeability in diabetic mice for several months following a single intraocular injection, accumulation of acriflavine in the retina resulted in retinal toxicity over time, raising concerns for its use in patients. Conversely, 32-134D, a recently developed HIF inhibitor structurally unrelated to acriflavine, was not toxic to the retina, yet effectively inhibited HIF accumulation and normalized HIF-regulated gene expression in mice and in human retinal organoids. Intraocular administration of 32-134D prevented retinal neovascularization and vascular hyperpermeability in mice. These results provide the foundation for clinical studies assessing 32-134D for the treatment of patients with diabetic eye disease.
Collapse
Affiliation(s)
- Jing Zhang
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Deepti Sharma
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aumreetam Dinabandhu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology and Diagnostic Sciences, School of Dentistry, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, USA
| | - Jaron Sanchez
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brooks Applewhite
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathleen Jee
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Monika Deshpande
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Miguel Flores-Bellver
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ming-Wen Hu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chuanyu Guo
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shaima Salman
- Armstrong Oxygen Biology Research Center; Vascular Program, Institute for Cell Engineering; Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, Biological Chemistry, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yousang Hwang
- Armstrong Oxygen Biology Research Center; Vascular Program, Institute for Cell Engineering; Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, Biological Chemistry, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicole M. Anders
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology and the Division of Clinical Pharmacology at the School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle A. Rudek
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology and the Division of Clinical Pharmacology at the School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jiang Qian
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - M. Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Gregg L. Semenza
- Armstrong Oxygen Biology Research Center; Vascular Program, Institute for Cell Engineering; Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, Biological Chemistry, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Silvia Montaner
- Department of Oncology and Diagnostic Sciences, School of Dentistry, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, USA
| | - Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
38
|
Cheng F, He J, Yang J. Bone marrow microenvironment: roles and therapeutic implications in obesity-associated cancer. Trends Cancer 2023; 9:566-577. [PMID: 37087397 PMCID: PMC10329995 DOI: 10.1016/j.trecan.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/24/2023]
Abstract
Obesity is increasing globally and has been closely linked to the initiation and progression of multiple human cancers. These relationships, to a large degree, are mediated through obesity-driven disruption of physiological homeostasis characterized by local and systemic endocrinologic, inflammatory, and metabolic changes. Bone marrow microenvironment (BMME), which evolves during obesity, has been implicated in multiple types of cancer. Growing evidence shows that physiological dysfunction of BMME with altered cellular composition, stromal and immune cell function, and energy metabolism, as well as inflammation and hypoxia, in the context of obesity contributes to cancer initiation and progression. Nonetheless, the mechanisms underlying the obesity-BMME-cancer axis remain elusive. In this review, we discuss the recent advances in understanding the evolution of BMME during obesity, its contributions to cancer initiation and progression, and the implications for cancer therapy.
Collapse
Affiliation(s)
- Feifei Cheng
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Jin He
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Jing Yang
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
39
|
Suresh MV, Aggarwal V, Raghavendran K. The Intersection of Pulmonary Vascular Disease and Hypoxia-Inducible Factors. Interv Cardiol Clin 2023; 12:443-452. [PMID: 37290846 DOI: 10.1016/j.iccl.2023.03.010] [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: 06/10/2023]
Abstract
Hypoxia-inducible factors (HIFs) are a family of nuclear transcription factors that serve as the master regulator of the adaptive response to hypoxia. In the lung, HIFs orchestrate multiple inflammatory pathways and signaling. They have been reported to have a major role in the initiation and progression of acute lung injury, chronic obstructive pulmonary disease, pulmonary fibrosis, and pulmonary hypertension. Although there seems to be a clear mechanistic role for both HIF 1α and 2α in pulmonary vascular diseases including PH, a successful translation into a definitive therapeutic modality has not been accomplished to date.
Collapse
Affiliation(s)
| | - Vikas Aggarwal
- Division of Cardiology (Frankel Cardiovascular Center), Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Section of Cardiology, Department of Internal Medicine, Veterans Affairs Medical Center, Ann Arbor, MI, USA
| | - Krishnan Raghavendran
- Division of Acute Care Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
40
|
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Understanding the relationship between cancer associated cachexia and hypoxia-inducible factor-1. Biomed Pharmacother 2023; 163:114802. [PMID: 37146421 DOI: 10.1016/j.biopha.2023.114802] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a multifactorial disorder characterized by an unrestricted loss of body weight as a result of muscle and adipose tissue atrophy. Cachexia is influenced by several factors, including decreased metabolic activity and food intake, an imbalance between energy uptake and expenditure, excessive catabolism, and inflammation. Cachexia is highly associated with all types of cancers responsible for more than half of cancer-related mortalities worldwide. In healthy individuals, adipose tissue significantly regulates energy balance and glucose homeostasis. However, in metastatic cancer patients, CAC occurs mainly because of an imbalance between muscle protein synthesis and degradation which are organized by certain extracellular ligands and associated signaling pathways. Under hypoxic conditions, hypoxia-inducible factor-1 (HIF-1α) accumulated and translocated to the nucleus and activate numerous genes involved in cell survival, invasion, angiogenesis, metastasis, metabolic reprogramming, and cancer stemness. On the other hand, the ubiquitination proteasome pathway is inhibited during low O2 levels which promote muscle wasting in cancer patients. Therefore, understanding the mechanism of the HIF-1 pathway and its metabolic adaptation to biomolecules is important for developing a novel therapeutic method for cancer and cachexia therapy. Even though many HIF inhibitors are already in a clinical trial, their mechanism of action remains unknown. With this background, this review summarizes the basic concepts of cachexia, the role of inflammatory cytokines, pathways connected with cachexia with special reference to the HIF-1 pathway and its regulation, metabolic changes, and inhibitors of HIFs.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
41
|
Jamshed F, Dashti F, Ouyang X, Mehal WZ, Banini BA. New uses for an old remedy: Digoxin as a potential treatment for steatohepatitis and other disorders. World J Gastroenterol 2023; 29:1824-1837. [PMID: 37032732 PMCID: PMC10080697 DOI: 10.3748/wjg.v29.i12.1824] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/12/2023] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
Repurposing of the widely available and relatively cheap generic cardiac gly-coside digoxin for non-cardiac indications could have a wide-ranging impact on the global burden of several diseases. Over the past several years, there have been significant advances in the study of digoxin pharmacology and its potential non-cardiac clinical applications, including anti-inflammatory, antineoplastic, metabolic, and antimicrobial use. Digoxin holds promise in the treatment of gastrointestinal disease, including nonalcoholic steatohepatitis and alcohol-associated steatohepatitis as well as in obesity, cancer, and treatment of viral infections, among other conditions. In this review, we provide a summary of the clinical uses of digoxin to date and discuss recent research on its emerging applications.
Collapse
Affiliation(s)
- Fatima Jamshed
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
- Griffin Hospital-Yale University, Derby, CT 06418, United States
| | - Farzaneh Dashti
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| | - Xinshou Ouyang
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
- West Haven Veterans Medical Center, West Haven, CT 06516, United States
| | - Bubu A Banini
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| |
Collapse
|
42
|
Yao X, Hu Q, Liu X, Ling Q, Leng Y, Zhao H, Yu P, Ma J, Zhao Y, Liu M, Yang R. Atrial fibrillation and breast cancer—Vicious twins? A systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1113231. [PMID: 36970342 PMCID: PMC10036368 DOI: 10.3389/fcvm.2023.1113231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/13/2023] [Indexed: 03/12/2023] Open
Abstract
BackgroundEpidemiological studies suggest a bidirectional association between atrial fibrillation and breast cancer. This study aimed to conduct a meta-analysis to elucidate the prevalence of atrial fibrillation among breast cancer patients, and the bidirectional association between atrial fibrillation and breast cancer.MethodsPubMed, the Cochrane Library, and Embase were searched to identify studies reporting the prevalence, incidence, and bidirectional association between atrial fibrillation and breast cancer. The study was registered with PROSPERO (CRD42022313251). Levels of evidence and recommendations were assessed by the Grading of Recommendations Assessment, Development and Evaluation (GRADE).ResultsTwenty-three studies (17 retrospective cohort studies, 5 case-control studies and 1 cross-sectional study) involving 8,537,551 participants were included. Among patients with breast cancer, the prevalence of atrial fibrillation was 3% (11 studies; 95% CI: 0.6 to 7.1%) and the incidence was 2.7% (6 studies; 95% CI: 1.1 to 4.9%). Breast cancer was associated with increased risk of atrial fibrillation (5 studies; hazard ratio [HR]: 1.43, 95% CI: 1.12 to 1.82, I2 = 98%). Atrial fibrillation was also significantly associated elevated risk of breast cancer (5 studies HR: 1.18, 95% CI: 1.14 to 1.22, I2 = 0%). Grade assessment shown low certainty of the evidence for the risk of atrial fibrillation and moderate certainty of the evidence for the risk of breast cancer.ConclusionAtrial fibrillation is not uncommon in patients with breast cancer and vice versa. There is a bidirectional association between atrial fibrillation (low certainty) and breast cancer (moderate certainty).
Collapse
Affiliation(s)
- Xiongda Yao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qingwen Hu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China
| | - Qing Ling
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yurong Leng
- The Affiliated Stomatological Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Huilei Zhao
- Department of Anesthesiology, The Third Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Peng Yu
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Yujie Zhao
- Department of Cardiology, Seventh People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Menglu Liu
- Department of Cardiology, Seventh People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Renqiang Yang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- *Correspondence: Renqiang Yang
| |
Collapse
|
43
|
Joe NS, Wang Y, Oza HH, Godet I, Milki N, Riggins GJ, Gilkes DM. Mebendazole Treatment Disrupts the Transcriptional Activity of Hypoxia-Inducible Factors 1 and 2 in Breast Cancer Cells. Cancers (Basel) 2023; 15:cancers15041330. [PMID: 36831670 PMCID: PMC9954103 DOI: 10.3390/cancers15041330] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Breast cancer is the most diagnosed cancer in women in the world. Mebendazole (MBZ) has been demonstrated to have preclinical efficacy across multiple cancers, including glioblastoma multiforme, medulloblastoma, colon, breast, pancreatic, and thyroid cancers. MBZ was also well tolerated in a recent phase I clinical trial of adults diagnosed with glioma. The mechanisms of action reported so far for MBZ include tubulin disruption, inhibiting angiogenesis, promoting apoptosis, and maintaining stemness. To elucidate additional mechanisms of action for mebendazole (MBZ), we performed RNA sequencing of three different breast cancer cell lines treated with either MBZ or vehicle control. We compared the top genes downregulated upon MBZ treatment with expression profiles of cells treated with over 15,000 perturbagens using the clue.io online analysis tool. In addition to tubulin inhibitors, the gene expression profile that correlated most with MBZ treatment matched the profile of cells treated with known hypoxia-inducible factor (HIF-1α and -2α) inhibitors. The HIF pathway is the main driver of the cellular response to hypoxia, which occurs in solid tumors. Preclinical data support using HIF inhibitors in combination with standard of care to treat solid tumors. Therefore, we tested the hypothesis that MBZ could inhibit the hypoxia response. Using RNA sequencing and HIF-reporter assays, we demonstrate that MBZ inhibits the transcriptional activity of HIFs in breast cancer cell lines and in mouse models of breast cancer by preventing the induction of HIF-1α, HIF-2α, and HIF-1β protein under hypoxia. Taken together, our results suggest that MBZ treatment has additional therapeutic efficacy in the setting of hypoxia and warrants further consideration as a cancer therapy.
Collapse
Affiliation(s)
- Natalie S. Joe
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuanfeng Wang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Biochemistry and Molecular Biology Program, The Johns Hopkins University School of Public Health, Baltimore, MD 21205, USA
| | - Harsh H. Oza
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Inês Godet
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nubaira Milki
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gregory J. Riggins
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Daniele M. Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Correspondence:
| |
Collapse
|
44
|
Trachtenberg B, Cowger J. HFSA Expert Consensus Statement on the Medical Management of Patients on Durable Mechanical Circulatory Support. J Card Fail 2023; 29:479-502. [PMID: 36828256 DOI: 10.1016/j.cardfail.2023.01.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 02/24/2023]
Abstract
The medical management of patients supported with durable continuous flow left ventricular assist device (LVAD) support encompasses pharmacologic therapies administered in the preoperative, intraoperative, postoperative and chronic LVAD support stages. As patients live longer on LVAD support, the risks of LVAD-related complications and progression of cardiovascular and other diseases increase. Using existing data from cohort studies, registries, randomized trials and expert opinion, this Heart Failure Society of America Consensus Document on the Medical Management of Patients on Durable Mechanical Circulatory Support offers best practices on the management of patients on durable MCS, focusing on pharmacological therapies administered to patients on continuous flow LVADs. While quality data in the LVAD population are few, the utilization of guideline directed heart failure medical therapies (GDMT) and the importance of blood pressure management, right ventricular preload and afterload optimization, and antiplatelet and anticoagulation regimens are discussed. Recommended pharmacologic regimens used to mitigate or treat common complications encountered during LVAD support, including arrhythmias, vasoplegia, mucocutaneous bleeding, and infectious complications are addressed. Finally, this document touches on important potential pharmacological interactions from anti-depressants, herbal and nutritional supplements of relevance to providers of patients on LVAD support.
Collapse
Affiliation(s)
- Barry Trachtenberg
- Houston Methodist Heart and Vascular Center, Methodist J.C. Walter Transplant Center.
| | - Jennifer Cowger
- Medical Director, Mechanical Circulatory Support Program, Codirector, Cardiac Critical Care, Henry Ford Advanced Heart Failure Program.
| |
Collapse
|
45
|
Guo C, Deshpande M, Niu Y, Kachwala I, Flores-Bellver M, Megarity H, Nuse T, Babapoor-Farrokhran S, Ramada M, Sanchez J, Inamdar N, Johnson TV, Canto-Soler MV, Montaner S, Sodhi A. HIF-1α accumulation in response to transient hypoglycemia may worsen diabetic eye disease. Cell Rep 2023; 42:111976. [PMID: 36640318 PMCID: PMC9960808 DOI: 10.1016/j.celrep.2022.111976] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 10/16/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Tight glycemic control (TGC), the cornerstone of diabetic management, reduces the incidence and progression of diabetic microvascular disease. However, TGC can also lead to transient episodes of hypoglycemia, which have been associated with adverse outcomes in patients with diabetes. Here, we demonstrate that low glucose levels result in hypoxia-inducible factor (HIF)-1-dependent expression of the glucose transporter, Glut1, in retinal cells. Enhanced nuclear accumulation of HIF-1α was independent of its canonical post-translational stabilization but instead dependent on stimulation of its translation and nuclear localization. In the presence of hypoxia, this physiologic response to low glucose resulted in a marked increase in the secretion of the HIF-dependent vasoactive mediators that promote diabetic retinopathy. Our results provide a molecular explanation for how early glucose control, as well as glycemic variability (i.e., oscillating serum glucose levels), contributes to diabetic eye disease. These observations have important implications for optimizing glucose management in patients with diabetes.
Collapse
Affiliation(s)
- Chuanyu Guo
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Monika Deshpande
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yueqi Niu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Isha Kachwala
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Miguel Flores-Bellver
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Haley Megarity
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Taylor Nuse
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Michael Ramada
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jaron Sanchez
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Neelay Inamdar
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Thomas V Johnson
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Maria Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Silvia Montaner
- Department of Oncology and Diagnostic Sciences, School of Dentistry, Greenebaum Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
46
|
Kim AH, Kolesnikova M, Ngo WK, Tsang SH. Effects of medications on hypoxia-inducible factor in the retina: A review. Clin Exp Ophthalmol 2023; 51:205-216. [PMID: 36594241 DOI: 10.1111/ceo.14161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023]
Abstract
Hypoxia-inducible factor (HIF) plays a critical role in the mechanisms that allow cells to adapt to various oxygen levels in the environment. Specifically, HIF-1⍺ has shown to be widely involved in cellular repair, survival, and energy metabolism. HIF-1⍺ has also been found in increased levels in cancer cells, highlighting the importance of balance in the hypoxic response. Promoting HIF-1⍺ activity as a potential therapy for degenerative diseases and inhibiting HIF-1⍺ as a therapy for pathologies with overactive cell proliferation are actively being explored. Digoxin and metformin, HIF-1⍺ inhibitors, and deferoxamine and ⍺-ketoglutarate analogues, HIF-1⍺ activators, are being studied for application in age-related macular degeneration, diabetic retinopathy, and retinitis pigmentosa. However, these same medications have retinal toxicities that must be assessed before implementation of therapeutic care. Herein, we highlight the duality of therapeutic and toxic potential of HIF-1⍺ that must be carefully assessed prior to its clinical application in retinal disorders.
Collapse
Affiliation(s)
- Angela H Kim
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,SUNY Downstate Medical School, Brooklyn, New York, USA
| | - Masha Kolesnikova
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,SUNY Downstate Medical School, Brooklyn, New York, USA
| | - Wei Kiong Ngo
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,Departments of Pathology & Cell Biology, Columbia Stem Cell Initiative, New York, New York, USA.,Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
47
|
Pamies D, Vujić T, Schvartz D, Boccard J, Repond C, Nunes C, Rudaz S, Sanchez JC, González-Ruiz V, Zurich MG. Digoxin Induces Human Astrocyte Reaction In Vitro. Mol Neurobiol 2023; 60:84-97. [PMID: 36223047 PMCID: PMC9758102 DOI: 10.1007/s12035-022-03057-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/21/2022] [Indexed: 12/30/2022]
Abstract
Astrocyte reaction is a complex cellular process involving astrocytes in response to various types of CNS injury and a marker of neurotoxicity. It has been abundantly studied in rodents but relatively poorly in human cells due to limited access to the brain. Astrocytes play important roles in cerebral energy metabolism and are also key players in neuroinflammation. Astroglial metabolic and inflammatory changes have been reported with age, leading to the hypothesis that mitochondrial metabolism and inflammatory responses are interconnected. However, the relationship between energy metabolism and astrocyte reactivity in the context of neurotoxicity is not known. We hypothesized that changes in energy metabolism of astrocytes will be coupled to their activation by xenobiotics. Astrocyte reaction and associated energy metabolic changes were assessed by immunostaining, gene expression, proteomics, metabolomics, and extracellular flux analyses after 24 h of exposure of human ReN-derived astrocytes to digoxin (1-10 µM) or TNFα (30 ng/ml) used as a positive control. Strong astrocytic reaction was observed, accompanied by increased glycolysis at low concentrations of digoxin (0.1 and 0.5 µM) and after TNFα exposure, suggesting that increased glycolysis may be a common feature of reactive astrocytes, independent of the triggering molecule. In conclusion, whether astrocyte activation is triggered by cytokines or a xenobiotic, it is strongly tied to energy metabolism in human ReN-derived astrocytes. Increased glycolysis might be considered as an endpoint to detect astrocyte activation by potentially neurotoxic compounds in vitro. Finally, ReN-derived astrocytes may help to decipher mechanisms of neurotoxicity in ascertaining the ability of chemicals to directly target astrocytes.
Collapse
Affiliation(s)
- David Pamies
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Tatjana Vujić
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Domitille Schvartz
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Julien Boccard
- Translational Biomarker Group, Department of Medicine, University of Geneva, Geneva, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Cendrine Repond
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland
| | - Carolina Nunes
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Serge Rudaz
- Translational Biomarker Group, Department of Medicine, University of Geneva, Geneva, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Víctor González-Ruiz
- Translational Biomarker Group, Department of Medicine, University of Geneva, Geneva, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Marie-Gabrielle Zurich
- Department of Biological Sciences, University of Lausanne, Lausanne, Switzerland ,School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
48
|
Zhang J, Hu Z, Horta CA, Yang J. Regulation of epithelial-mesenchymal transition by tumor microenvironmental signals and its implication in cancer therapeutics. Semin Cancer Biol 2023; 88:46-66. [PMID: 36521737 DOI: 10.1016/j.semcancer.2022.12.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been implicated in various aspects of tumor development, including tumor invasion and metastasis, cancer stemness, and therapy resistance. Diverse stroma cell types along with biochemical and biophysical factors in the tumor microenvironment impinge on the EMT program to impact tumor progression. Here we provide an in-depth review of various tumor microenvironmental signals that regulate EMT in cancer. We discuss the molecular mechanisms underlying the role of EMT in therapy resistance and highlight new therapeutic approaches targeting the tumor microenvironment to impact EMT and tumor progression.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Zhimin Hu
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Calista A Horta
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
49
|
Yang Y, Chen C, Zuo Q, Lu H, Salman S, Lyu Y, Huang TYT, Wicks EE, Jackson W, Datan E, Wang R, Wang Y, Le N, Zhu Y, Qin W, Semenza GL. NARF is a hypoxia-induced coactivator for OCT4-mediated breast cancer stem cell specification. SCIENCE ADVANCES 2022; 8:eabo5000. [PMID: 36490339 PMCID: PMC9733926 DOI: 10.1126/sciadv.abo5000] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/27/2022] [Indexed: 06/17/2023]
Abstract
Hypoxia is a key characteristic of the breast cancer microenvironment that promotes expression of the transcriptional activator hypoxia-inducible factor 1 (HIF-1) and is associated with poor patient outcome. HIF-1 increases the expression or activity of stem cell pluripotency factors, which control breast cancer stem cell (BCSC) specification and are required for cancer metastasis. Here, we identify nuclear prelamin A recognition factor (NARF) as a hypoxia-inducible, HIF-1 target gene in human breast cancer cells. NARF functions as an essential coactivator by recruiting the histone demethylase KDM6A to OCT4 bound to genes encoding the pluripotency factors NANOG, KLF4, and SOX2, leading to demethylation of histone H3 trimethylated at lysine-27 (H3K27me3), thereby increasing the expression of NANOG, KLF4, and SOX2, which, together with OCT4, mediate BCSC specification. Knockdown of NARF significantly decreased the BCSC population in vitro and markedly impaired tumor initiation capacity and lung metastasis in orthotopic mouse models.
Collapse
Affiliation(s)
- Yongkang Yang
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21205, USA
| | - Chelsey Chen
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qiaozhu Zuo
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Haiquan Lu
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21205, USA
| | - Shaima Salman
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yajing Lyu
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tina Yi-Ting Huang
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth E. Wicks
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Walter Jackson
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emmanuel Datan
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ru Wang
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yufeng Wang
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nguyet Le
- Predoctoral Training Program in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yayun Zhu
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Gregg L. Semenza
- Armstrong Oxygen Biology Research Center and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21205, USA
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
50
|
Ainembabazi D, Geng X, Gavande NS, Turchi JJ, Zhang Y. Synthesis and Biological Evaluation of Cardiac Glycosides for Cancer Therapy by Targeting the DNA Damage Response. ChemMedChem 2022; 17:e202200415. [PMID: 36054918 PMCID: PMC9637767 DOI: 10.1002/cmdc.202200415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/29/2022] [Indexed: 01/12/2023]
Abstract
Cardiac glycosides (CGs) are bioactive compounds originally used to treat heart diseases, but recent studies have demonstrated their anticancer activity. We previously demonstrated that Antiaris toxicaria 2 (AT2) possesses anticancer activity in KRAS mutated lung cancers via impinging on the DNA damage response (DDR) pathway. Toward developing this class of molecules for cancer therapy, herein we report a multistep synthetic route utilizing k-strophanthidin as the initial building block for determination of structure-activity relationships (SARs). A systematic structural design approach was applied that included modifications of the sugar moiety, the glycoside linker, stereochemistry, and lactone ring substitutions to generate a library of O-glycosides and MeON-neoglycosides derivatives. These molecules were screened for their anticancer activities and their impact on DDR signaling in KRAS mutant lung cancer cells. These results demonstrate the ability to chemically synthesize CG derivatives and define the SARs to optimize AT2 as a cancer therapeutic.
Collapse
Affiliation(s)
- Diana Ainembabazi
- Department of MedicineHematology/OncologyIndiana UniversitySchool of Medicine980 W. Walnut Street, C56046202IndianapolisINUSA
| | - Xinran Geng
- Department of PharmacologyCase Western Reserve UniversitySchool of Medicine10900 Euclid Avenue44106ClevelandOHUSA
| | - Navnath S. Gavande
- Department of Pharmaceutical SciencesEugene Applebaum College of Pharmacy and Health SciencesWayne State University259 Mack Avenue48201DetroitMIUSA,Molecular Therapeutics ProgramBarbara Ann Karmanos Cancer InstituteWayne State University4100 John R48201DetroitMIUSA
| | - John J. Turchi
- Department of MedicineHematology/OncologyIndiana UniversitySchool of Medicine980 W. Walnut Street, C56046202IndianapolisINUSA
| | - Youwei Zhang
- Department of PharmacologyCase Western Reserve UniversitySchool of Medicine10900 Euclid Avenue44106ClevelandOHUSA
| |
Collapse
|