1
|
Kong F, Lei L, Cai L, Li J, Zhao C, Liu M, Qi D, Gao J, Li E, Gao W, Du X, Song Y, Liu G, Li X. Hypoxia-inducible factor 2α mediates nonesterified fatty acids and hypoxia-induced lipid accumulation in bovine hepatocytes. J Dairy Sci 2025; 108:4062-4078. [PMID: 39890076 DOI: 10.3168/jds.2024-25839] [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: 10/08/2024] [Accepted: 12/23/2024] [Indexed: 02/03/2025]
Abstract
Ketosis is a metabolic disorder frequently occurring in the perinatal period, characterized by elevated circulating concentrations of nonesterified fatty acids (NEFA) due to negative energy balance, resulting in fatty liver in dairy cows. However, the mechanism of hepatic steatosis induced by high concentrations of NEFA in ketosis remains unclear. Hypoxia-inducible factor 2α (HIF-2α), which mediates adaptation to hypoxic stress, plays a critical role in regulating lipid metabolism. In this study, we investigate whether HIF-2α is involved in NEFA-driven hepatic lipid accumulation in dairy cows with ketosis. Liver and blood samples were collected from 10 healthy cows (blood BHB concentration <1.2 mM) and 10 ketotic cows (blood BHB concentration >3.0 mM with clinical symptoms) with similar lactation numbers (median = 3, range = 2-4) at 3 to 9 DIM (median = 6). In cows with ketosis, serum concentrations of NEFA and BHB were greater, but serum concentrations of glucose were lower. Moreover, hepatic triglyceride content increased significantly. In the liver of ketotic cows, which was accompanied by upregulated HIF-2α expression. To determine the potential association among hypoxia, HIF-2α, and the formation of hepatocellular steatosis in vitro, we isolated hepatocytes from healthy calves for the following experiments. First, hepatocytes were treated with 0, 0.6, 1.2, or 2.4 mM NEFA (52.7 mM stock NEFA solution was diluted in RPMI-1640 basic medium supplemented with 2% fatty acid-free BSA to achieve the specified concentrations) for 18 h, showing that HIF-2α expression and cellular hypoxia occurred in a dose-dependent manner. Next, hepatocytes were infected with HIF-2α (encoded by EPAS1) small interfering RNA (Si-HIF-2α) for 48 h and then treated with 1.2 mM NEFA for 18 h. Results indicated that silencing HIF-2α decreased NEFA-induced lipid accumulation in bovine hepatocytes. Subsequently, hepatocytes treated with or without NEFA were placed in an AnaeroPack System, mimicking a hypoxic condition, for 0, 12, 18, or 24 h. Results showed that hypoxia could induce and further exacerbate lipid accumulation in bovine hepatocytes. Meanwhile, normal or NEFA-treated hepatocytes were cocultured with or without PT2385, a specific HIF-2α inhibitor, showing that hypoxia promoted steatosis through HIF-2α. Activating transcription factor 4 (ATF4) is an endoplasmic reticulum (ER) stress and hypoxia-inducible transcription factor. Here, bovine hepatocytes were treated with NEFA or hypoxia following transfecting ATF4 small interfering RNA, which demonstrated that ATF4 knockdown alleviated the extent of lipid accumulation in bovine hepatocytes. In addition, we found that ATF4 expression was correlated with HIF-2α levels in both liver tissue and cultured hepatocyte models. Moreover, overexpression of ATF4 weakened the beneficial effects of HIF-2α inhibition. Overall, these data suggest that NEFA-induced hepatic hypoxia significantly contributes to the progression of hepatic steatosis which in turn, intensifies hypoxia and leads to a self-perpetuating cycle of reciprocal causation, further exacerbating hepatic lipid deposition. Additionally, accumulated HIF-2α plays a critical role in this complex-origin steatosis, potentially through ATF4.
Collapse
Affiliation(s)
- Fanrong Kong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Cai
- College of Food and Biology of Changchun Polytechnic, Changchun 130062, China
| | - Jinxia Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Chenchen Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Menglin Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Dandan Qi
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jie Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Enzhu Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiliang Du
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuxiang Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guowen Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
2
|
Solanki S, Shah YM. Hypoxia-Induced Signaling in Gut and Liver Pathobiology. ANNUAL REVIEW OF PATHOLOGY 2024; 19:291-317. [PMID: 37832943 DOI: 10.1146/annurev-pathmechdis-051122-094743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Oxygen (O2) is essential for cellular metabolism and biochemical reactions. When the demand for O2 exceeds the supply, hypoxia occurs. Hypoxia-inducible factors (HIFs) are essential to activate adaptive and survival responses following hypoxic stress. In the gut (intestines) and liver, the presence of oxygen gradients or physiologic hypoxia is necessary to maintain normal homeostasis. While physiologic hypoxia is beneficial and aids in normal functions, pathological hypoxia is harmful as it exacerbates inflammatory responses and tissue dysfunction and is a hallmark of many cancers. In this review, we discuss the role of gut and liver hypoxia-induced signaling, primarily focusing on HIFs, in the physiology and pathobiology of gut and liver diseases. Additionally, we examine the function of HIFs in various cell types during gut and liver diseases, beyond intestinal epithelial and hepatocyte HIFs. This review highlights the importance of understanding hypoxia-induced signaling in the pathogenesis of gut and liver diseases and emphasizes the potential of HIFs as therapeutic targets.
Collapse
Affiliation(s)
- Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Ahire D, Mariasoosai C, Naji-Talakar S, Natesan S, Prasad B. Promiscuity and Quantitative Contribution of UGT2B17 in Drug and Steroid Metabolism Determined by Experimental and Computational Approaches. J Chem Inf Model 2024; 64:483-498. [PMID: 38198666 DOI: 10.1021/acs.jcim.3c01514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Uridine 5'-diphospho-glulcuronosyltransferase 2B17 (UGT2B17) is important in the metabolism of steroids and orally administered drugs due to its high interindividual variability. However, the structural basis governing the substrate selectivity or inhibition of UGT2B17 remains poorly understood. This study investigated 76 FDA-approved drugs and 20 steroids known to undergo glucuronidation for their metabolism by UGT2B17. Specifically, we assessed the substrate selectivity for UGT2B17 over other UGT enzymes using recombinant human UGT2B17 (rUGT2B17), human intestinal microsomes, and human liver microsomes. The quantitative contribution of intestinal UGT2B17 in the glucuronidation of these compounds was characterized using intestinal microsomes isolated from UGT2B17 expressors and nonexpressors. In addition, a structure-based pharmacophore model for UGT2B17 substrates was built and validated using the studied pool of substrates and nonsubstrates. The results show that UGT2B17 could metabolize 23 out of 96 compounds from various chemical classes, including alcohols and carboxylic acids, particularly in the intestine. Interestingly, amines were less susceptible to UGT2B17 metabolism, though they could inhibit the enzyme. Three main pharmacophoric features of UGT2B17 substrates include (1) the presence of an accessible -OH or -COOH group near His35 residue, (2) a hydrophobic functional group at ∼4.5-5 Å from feature 1, and (3) an aromatic ring ∼5-7 Å from feature 2. Most of the studied compounds inhibited UGT2B17 activity irrespective of their substrate potential, indicating the possibility of multiple mechanisms. These data suggest that UGT2B17 is promiscuous in substrate selectivity and inhibition and has a high potential to produce significant variability in the absorption and disposition of orally administered drugs.
Collapse
Affiliation(s)
- Deepak Ahire
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Charles Mariasoosai
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Siavosh Naji-Talakar
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Senthil Natesan
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| |
Collapse
|
4
|
Strowd R, Ellingson B, Raymond C, Yao J, Wen PY, Ahluwalia M, Piotrowski A, Desai A, Clarke JL, Lieberman FS, Desideri S, Nabors LB, Ye X, Grossman S. Activity of a first-in-class oral HIF2-alpha inhibitor, PT2385, in patients with first recurrence of glioblastoma. J Neurooncol 2023; 165:101-112. [PMID: 37864646 PMCID: PMC10863646 DOI: 10.1007/s11060-023-04456-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/23/2023]
Abstract
INTRODUCTION Hypoxia inducible factor 2-alpha (HIF2α) mediates cellular responses to hypoxia and is over-expressed in glioblastoma (GBM). PT2385 is an oral HIF2α inhibitor with in vivo activity against GBM. METHODS A two-stage single-arm open-label phase II study of adults with GBM at first recurrence following chemoradiation with measurable disease was conducted through the Adult Brain Tumor Consortium. PT2385 was administered at the phase II dose (800 mg b.i.d.). The primary outcome was objective radiographic response (ORR = complete response + partial response, CR + PR); secondary outcomes were safety, overall survival (OS), and progression free survival (PFS). Exploratory objectives included pharmacokinetics (day 15 Cmin), pharmacodynamics (erythropoietin, vascular endothelial growth factor), and pH-weighted amine- chemical exchange saturation transfer (CEST) MRI to quantify tumor acidity at baseline and explore associations with drug response. Stage 1 enrolled 24 patients with early stoppage for ≤ 1 ORR. RESULTS Of the 24 enrolled patients, median age was 62.1 (38.7-76.7) years, median KPS 80, MGMT promoter was methylated in 46% of tumors. PT2385 was well tolerated. Grade ≥ 3 drug-related adverse events were hypoxia (n = 2), hyponatremia (2), lymphopenia (1), anemia (1), and hyperglycemia (1). No objective radiographic responses were observed; median PFS was 1.8 months (95% CI 1.6-2.5) and OS was 7.7 months (95% CI 4.9-12.6). Drug exposure varied widely and did not differ by corticosteroid use (p = 0.12), antiepileptics (p = 0.09), or sex (p = 0.37). Patients with high systemic exposure had significantly longer PFS (6.7 vs 1.8 months, p = 0.009). Baseline acidity by pH-weighted CEST MRI correlated significantly with treatment duration (R2 = 0.49, p = 0.017). Non-enhancing infiltrative disease with high acidity gave rise to recurrence. CONCLUSIONS PT2385 monotherapy had limited activity in first recurrent GBM. Drug exposure was variable. Signals of activity were observed in GBM patients with high systemic exposure and acidic lesions on CEST imaging. A second-generation HIF2α inhibitor is being studied.
Collapse
Affiliation(s)
- Roy Strowd
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston Salem, NC, 27104, USA.
| | | | | | - Jingwen Yao
- University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | | - Arati Desai
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - L Burt Nabors
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaobu Ye
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - Stuart Grossman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
5
|
O'Hagan D. The Emergence and Properties of Selectively Fluorinated 'Janus' Cyclohexanes. CHEM REC 2023; 23:e202300027. [PMID: 37016509 DOI: 10.1002/tcr.202300027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/14/2023] [Indexed: 04/06/2023]
Abstract
This account describes the evolution of a research programme that started by linking fluoromethylene (-CHF-) groups along aliphatic chains and then progressing to alicyclic rings with contiguous fluorine atoms. Different stereoisomers of aliphatic chains tend to adopt low polarity conformations. In order to force polar conformations, the programme began to address ring systems and in particular cyclohexanes, to restrain conformational freedom and co-aligned C-F bonds. The flagship molecule, all-cis-1,2,3,4,5,6-hexafluorocyclohexane 7, emerged to be the most polar aliphatic compound recorded. The polarity arises because there are three co-aligned triaxial C-F bonds and the six fluorines occupy one face of the ring. Conversely the electropositive hydrogens occupy the other face. These have been termed Janus face cyclohexanes after the Roman god with two faces. The review outlines progress by our group and others in preparing derivatives of the parent cyclohexane 7, in order to explore properties and potential applications of these Janus cyclohexanes.
Collapse
Affiliation(s)
- David O'Hagan
- University of St Andrews, St. Andrews, United Kingdom
| |
Collapse
|
6
|
Ahire D, Heyward S, Prasad B. Intestinal Metabolism of Diclofenac by Polymorphic UGT2B17 Correlates with its Highly Variable Pharmacokinetics and Safety across Populations. Clin Pharmacol Ther 2023; 114:161-172. [PMID: 37042794 PMCID: PMC10330245 DOI: 10.1002/cpt.2907] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/21/2023] [Indexed: 04/13/2023]
Abstract
Although the United States and Europe have shifted to the prescription use of oral diclofenac due to several serious incidences of cardiotoxicity, it is one of the most commonly used over-the-counter (OTC) pain medicines in major parts of the world. We elucidated the quantitative and tissue-specific contribution of uridine diphosphate-glucuronosyltransferases 17 (UGT2B17) in diclofenac metabolism and pharmacokinetics (PK). UGT2B17 is one of most deleted genes in humans with the gene deletion frequency ranging from ~ 20% in White population to 90% in Japanese population. The human intestinal and liver microsomes isolated from the high-UGT2B17 expressing individuals showed 21- and 4-fold greater rate of diclofenac glucuronide (DG) formation than in the null-UGT2B17 carriers, respectively. The greater contribution of intestinal UGT2B17 was confirmed by a strong correlation (R = 0.78, P < 0.001) between UGT2B17 abundance and DG formation in individual intestinal microsomes (n = 14). However, because UGT2B17 is a minor UGT isoform in the liver, DG formation rate correlated better with the expression of UGT2B7. The proteomics-informed physiologically-based pharmacokinetic (PBPK) model explains the reported higher exposure of diclofenac in women consistent with ~ 3-fold lower expression of UGT2B17. Similarly, our in silico predictions also corroborate with the reported higher exposure and lower standard clinical dose of diclofenac in Japanese population. Therefore, variable UGT2B17 mediated metabolism of oral diclofenac is a cause of concern, especially in the developing countries where it is still used as an OTC drug. The ontogeny data of UGTs in human hepatocytes can be utilized in developing PBPK models for predicting PK in the pediatric population.
Collapse
Affiliation(s)
- Deepak Ahire
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | | | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| |
Collapse
|
7
|
Quintás G, Castell JV, Moreno-Torres M. The assessment of the potential hepatotoxicity of new drugs by in vitro metabolomics. Front Pharmacol 2023; 14:1155271. [PMID: 37214440 PMCID: PMC10196061 DOI: 10.3389/fphar.2023.1155271] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Drug hepatotoxicity assessment is a relevant issue both in the course of drug development as well as in the post marketing phase. The use of human relevant in vitro models in combination with powerful analytical methods (metabolomic analysis) is a promising approach to anticipate, as well as to understand and investigate the effects and mechanisms of drug hepatotoxicity in man. The metabolic profile analysis of biological liver models treated with hepatotoxins, as compared to that of those treated with non-hepatotoxic compounds, provides useful information for identifying disturbed cellular metabolic reactions, pathways, and networks. This can later be used to anticipate, as well to assess, the potential hepatotoxicity of new compounds. However, the applicability of the metabolomic analysis to assess the hepatotoxicity of drugs is complex and requires careful and systematic work, precise controls, wise data preprocessing and appropriate biological interpretation to make meaningful interpretations and/or predictions of drug hepatotoxicity. This review provides an updated look at recent in vitro studies which used principally mass spectrometry-based metabolomics to evaluate the hepatotoxicity of drugs. It also analyzes the principal drawbacks that still limit its general applicability in safety assessment screenings. We discuss the analytical workflow, essential factors that need to be considered and suggestions to overcome these drawbacks, as well as recent advancements made in this rapidly growing field of research.
Collapse
Affiliation(s)
- Guillermo Quintás
- Metabolomics and Bioanalysis, Health and Biomedicine, Leitat Technological Center, Barcelona, Spain
- Analytical Unit, Health Research Institute La Fe, Valencia, Spain
| | - José V. Castell
- Unidad Mixta de Hepatología Experimental, Instituto de Investigación Sanitaria del Hospital La Fe (IIS La Fe), Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- CIBEREHD, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Moreno-Torres
- Unidad Mixta de Hepatología Experimental, Instituto de Investigación Sanitaria del Hospital La Fe (IIS La Fe), Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- CIBEREHD, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Guo M, Niu Y, Xie M, Liu X, Li X. Notch signaling, hypoxia, and cancer. Front Oncol 2023; 13:1078768. [PMID: 36798826 PMCID: PMC9927648 DOI: 10.3389/fonc.2023.1078768] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Abstract
Notch signaling is involved in cell fate determination and deregulated in human solid tumors. Hypoxia is an important feature in many solid tumors, which activates hypoxia-induced factors (HIFs) and their downstream targets to promote tumorigenesis and cancer development. Recently, HIFs have been shown to trigger the Notch signaling pathway in a variety of organisms and tissues. In this review, we focus on the pro- and anti-tumorigenic functions of Notch signaling and discuss the crosstalk between Notch signaling and cellular hypoxic response in cancer pathogenesis, including epithelia-mesenchymal transition, angiogenesis, and the maintenance of cancer stem cells. The pharmacological strategies targeting Notch signaling and hypoxia in cancer are also discussed in this review.
Collapse
Affiliation(s)
- Mingzhou Guo
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yang Niu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Min Xie
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xiaochen Li
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China,*Correspondence: Xiaochen Li,
| |
Collapse
|
9
|
Wu Z, Kim GJ, Park SY, Shon JC, Liu KH, Choi H. In Vitro Metabolism Study of Seongsanamide A in Human Liver Microsomes Using Non-Targeted Metabolomics and Feature-Based Molecular Networking. Pharmaceutics 2021; 13:pharmaceutics13071031. [PMID: 34371722 PMCID: PMC8309059 DOI: 10.3390/pharmaceutics13071031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 11/16/2022] Open
Abstract
Seongsanamide A is a bicyclic peptide with an isodityrosine residue discovered in Bacillus safensis KCTC 12796BP which exhibits anti-allergic activity in vitro and in vivo without significant cytotoxicity. The purpose of this study was to elucidate the in vitro metabolic pathway and potential for drug interactions of seongsanamide A in human liver microsomes using non-targeted metabolomics and feature-based molecular networking (FBMN) techniques. We identified four metabolites, and their structures were elucidated by interpretation of high-resolution tandem mass spectra. The primary metabolic pathway associated with seongsanamide A metabolism was hydroxylation and oxidative hydrolysis. A reaction phenotyping study was also performed using recombinant cytochrome P450 isoforms. CYP3A4 and CYP3A5 were identified as the major metabolic enzymes responsible for metabolite formation. Seongsanamide A did not inhibit the cytochrome P450 isoforms commonly involved in drug metabolism (IC50 > 10 µM). These results will contribute to further understanding the metabolism and drug interaction potential of various bicyclic peptides.
Collapse
Affiliation(s)
- Zhexue Wu
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
| | - Geum Jin Kim
- College of Pharmacy and Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - So-Young Park
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (J.C.S.)
| | - Jong Cheol Shon
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (J.C.S.)
| | - Kwang-Hyeon Liu
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (J.C.S.)
- Correspondence: (K.-H.L.); (H.C.); Tel.: +82-53-950-8567 (K.-H.L.); +82-53-810-2824 (H.C.); FAX: +82- 53-950-8557 (K.-H.L.); +82-53-810-2036 (H.C.)
| | - Hyukjae Choi
- College of Pharmacy and Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
- Correspondence: (K.-H.L.); (H.C.); Tel.: +82-53-950-8567 (K.-H.L.); +82-53-810-2824 (H.C.); FAX: +82- 53-950-8557 (K.-H.L.); +82-53-810-2036 (H.C.)
| |
Collapse
|
10
|
Nwagwu CD, Adamson DC. Can we rely on synthetic pharmacotherapy for the treatment of glioblastoma? Expert Opin Pharmacother 2021; 22:1983-1994. [PMID: 34219576 DOI: 10.1080/14656566.2021.1950139] [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: 10/20/2022]
Abstract
Introduction: Despite decades of clinical trials utilizing conventional and novel therapeutics, the effective treatment of glioblastoma remains one of the most formidable challenges in oncology. Current standard of care includes surgery and chemoradiation. Synthetic pharmacotherapies continue to be explored as potential therapeutic options for glioblastoma patients.Areas covered: This study reviews synthetic pharmacotherapies that are currently under investigation in phase I-III clinical trials. The authors of this study highlight the mechanisms of action of the synthetic pharmacotherapy agents under investigation, outline the available evidence for their utility based on the literature, and summarize the current landscape.Expert opinion: Although warranting further investigation, the studies generally highlighted here have not shown remarkable changes in clinical benefits beyond what has already been established with radiochemotherapy. As we develop more synthetics, we will likely need to combine them with other synthetics to target multiple separate molecular pathways. There is considerable potential when this treatment strategy is guided by molecular profiling approaches which seek to stratify patients based on treatments that would be most efficacious for them.
Collapse
Affiliation(s)
- Chibueze D Nwagwu
- Department of Neurosurgery, Emory University, Atlanta, 30322-1007, United States
| | - David C Adamson
- Department of Neurosurgery, Emory University, Atlanta, 30322-1007, United States
| |
Collapse
|
11
|
Zhang Y, Coleman M, Brekken RA. Perspectives on Hypoxia Signaling in Tumor Stroma. Cancers (Basel) 2021; 13:3070. [PMID: 34202979 PMCID: PMC8234221 DOI: 10.3390/cancers13123070] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a well-known characteristic of solid tumors that contributes to tumor progression and metastasis. Oxygen deprivation due to high demand of proliferating cancer cells and standard of care therapies induce hypoxia. Hypoxia signaling, mainly mediated by the hypoxia-inducible transcription factor (HIF) family, results in tumor cell migration, proliferation, metabolic changes, and resistance to therapy. Additionally, the hypoxic tumor microenvironment impacts multiple cellular and non-cellular compartments in the tumor stroma, including disordered tumor vasculature, homeostasis of ECM. Hypoxia also has a multifaceted and often contradictory influence on immune cell function, which contributes to an immunosuppressive environment. Here, we review the important function of HIF in tumor stromal components and summarize current clinical trials targeting hypoxia. We provide an overview of hypoxia signaling in tumor stroma that might help address some of the challenges associated with hypoxia-targeted therapies.
Collapse
Affiliation(s)
- Yuqing Zhang
- Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX 75390, USA; (Y.Z.); (M.C.)
- Department of Surgery, UT Southwestern, Dallas, TX 75390, USA
- Cancer Biology Graduate Program, UT Southwestern, Dallas, TX 75390, USA
| | - Morgan Coleman
- Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX 75390, USA; (Y.Z.); (M.C.)
- Division of Pediatric Hematology and Oncology, UT Southwestern, Dallas, TX 75390, USA
| | - Rolf A. Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX 75390, USA; (Y.Z.); (M.C.)
- Department of Surgery, UT Southwestern, Dallas, TX 75390, USA
- Cancer Biology Graduate Program, UT Southwestern, Dallas, TX 75390, USA
| |
Collapse
|
12
|
Kim SE, Ji SB, Kim E, Jeong M, Kim J, Lee GM, Seo HJ, Bae S, Jeong Y, Lee S, Kim S, Lee T, Cho SJ, Liu KH. Nontargeted Metabolomics by High-Resolution Mass Spectrometry to Study the In Vitro Metabolism of a Dual Inverse Agonist of Estrogen-Related Receptors β and γ, DN203368. Pharmaceutics 2021; 13:pharmaceutics13060776. [PMID: 34072800 PMCID: PMC8230175 DOI: 10.3390/pharmaceutics13060776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
DN203368 ((E)-3-[1-(4-[4-isopropylpiperazine-1-yl]phenyl) 3-methyl-2-phenylbut-1-en-1-yl] phenol) is a 4-hydroxy tamoxifen analog that is a dual inverse agonist of estrogen-related receptor β/γ (ERRβ/γ). ERRγ is an orphan nuclear receptor that plays an important role in development and homeostasis and holds potential as a novel therapeutic target in metabolic diseases such as diabetes mellitus, obesity, and cancer. ERRβ is also one of the orphan nuclear receptors critical for many biological processes, such as development. We investigated the in vitro metabolism of DN203368 by conventional and metabolomic approaches using high-resolution mass spectrometry. The compound (100 μM) was incubated with rat and human liver microsomes in the presence of NADPH. In the metabolomic approach, the m/z value and retention time information obtained from the sample and heat-inactivated control group were statistically evaluated using principal component analysis and orthogonal partial least-squares discriminant analysis. Significant features responsible for group separation were then identified using tandem mass spectra. Seven metabolites of DN203368 were identified in rat liver microsomes and the metabolic pathways include hydroxylation (M1-3), N-oxidation (M4), N-deisopropylation (M5), N,N-dealkylation (M6), and oxidation and dehydrogenation (M7). Only five metabolites (M2, M3, and M5-M7) were detected in human liver microsomes. In the conventional approach using extracted ion monitoring for values of mass increase or decrease by known metabolic reactions, only five metabolites (M1-M5) were found in rat liver microsomes, whereas three metabolites (M2, M3, and M5) were found in human liver microsomes. This study revealed that nontargeted metabolomics combined with high-resolution mass spectrometry and multivariate analysis could be a more efficient tool for drug metabolite identification than the conventional approach. These results might also be useful for understanding the pharmacokinetics and metabolism of DN203368 in animals and humans.
Collapse
Affiliation(s)
- Sin-Eun Kim
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Seung-Bae Ji
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Euihyeon Kim
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Minseon Jeong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (M.J.); (J.K.)
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (M.J.); (J.K.)
| | - Gyung-Min Lee
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Hyung-Ju Seo
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Subin Bae
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Yeojin Jeong
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Sangkyu Lee
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
| | - Sunghwan Kim
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
- Department of Chemistry, Kyungpook National University, Daegu 41566, Korea
| | - Taeho Lee
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
| | - Sung Jin Cho
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, Korea
- Correspondence: (S.J.C.); (K.-H.L.); Tel.: +82-2-958-5137 (S.-J.C.); +82-53-950-8567 (K.-H.L.); Fax: +82-2-958-5137 (S.J.C.); +82-53-950-8557 (K.-H.L.)
| | - Kwang-Hyeon Liu
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-E.K.); (S.-B.J.); (E.K.); (G.-M.L.); (H.-J.S.); (S.B.); (Y.J.); (S.L.); (T.L.)
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea;
- Correspondence: (S.J.C.); (K.-H.L.); Tel.: +82-2-958-5137 (S.-J.C.); +82-53-950-8567 (K.-H.L.); Fax: +82-2-958-5137 (S.J.C.); +82-53-950-8557 (K.-H.L.)
| |
Collapse
|
13
|
Johnson BM, Shu YZ, Zhuo X, Meanwell NA. Metabolic and Pharmaceutical Aspects of Fluorinated Compounds. J Med Chem 2020; 63:6315-6386. [PMID: 32182061 DOI: 10.1021/acs.jmedchem.9b01877] [Citation(s) in RCA: 359] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The applications of fluorine in drug design continue to expand, facilitated by an improved understanding of its effects on physicochemical properties and the development of synthetic methodologies that are providing access to new fluorinated motifs. In turn, studies of fluorinated molecules are providing deeper insights into the effects of fluorine on metabolic pathways, distribution, and disposition. Despite the high strength of the C-F bond, the departure of fluoride from metabolic intermediates can be facile. This reactivity has been leveraged in the design of mechanism-based enzyme inhibitors and has influenced the metabolic fate of fluorinated compounds. In this Perspective, we summarize the literature associated with the metabolism of fluorinated molecules, focusing on examples where the presence of fluorine influences the metabolic profile. These studies have revealed potentially problematic outcomes with some fluorinated motifs and are enhancing our understanding of how fluorine should be deployed.
Collapse
Affiliation(s)
- Benjamin M Johnson
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Yue-Zhong Shu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb Company, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Xiaoliang Zhuo
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Nicholas A Meanwell
- Discovery Chemistry Platforms, Small Molecule Drug Discovery, Bristol Myers Squibb Company, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| |
Collapse
|
14
|
Zhang H, Basit A, Wolford C, Chen KF, Gaedigk A, Lin YS, Leeder JS, Prasad B. Normalized Testosterone Glucuronide as a Potential Urinary Biomarker for Highly Variable UGT2B17 in Children 7-18 Years. Clin Pharmacol Ther 2020; 107:1149-1158. [PMID: 31900930 DOI: 10.1002/cpt.1764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022]
Abstract
UDP-glucuronosyltransferase 2B17 (UGT2B17) is a highly variable androgen-metabolizing and drug-metabolizing enzyme. UGT2B17 exhibits a unique ontogeny profile characterized by a dramatic increase in hepatic protein expression from prepubertal age to adulthood. Age, sex, copy number variation (CNV), and single nucleotide polymorphisms only explain 26% of variability in protein expression, highlighting the need for a phenotypic biomarker for predicting interindividual variability in glucuronidation of UGT2B17 substrates. Here, we propose testosterone glucuronide (TG) normalized by androsterone glucuronide (TG/AG) as a urinary UGT2B17 biomarker, and examine the associations among urinary TG/AG and age, sex, and CNV. We performed targeted metabolomics of 12 androgen conjugates with liquid-chromatography tandem mass spectrometry in 63 pediatric subjects ages 7-18 years followed over 7 visits in 3 years. Consistent with the reported developmental trajectory of UGT2B17 protein expression, urinary TG/AG is significantly associated with age, sex, and CNV. In conclusion, TG/AG shows promise as a phenotypic urinary UGT2B17 biomarker.
Collapse
Affiliation(s)
- Haeyoung Zhang
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Abdul Basit
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Chris Wolford
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Kuan-Fu Chen
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Yvonne S Lin
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
15
|
Cannito S, Foglia B, Villano G, Turato C, C Delgado T, Morello E, Pin F, Novo E, Napione L, Quarta S, Ruvoletto M, Fasolato S, Zanus G, Colombatto S, Lopitz-Otsoa F, Fernández-Ramos D, Bussolino F, Sutti S, Albano E, Martínez-Chantar ML, Pontisso P, Parola M. SerpinB3 Differently Up-Regulates Hypoxia Inducible Factors -1α and -2α in Hepatocellular Carcinoma: Mechanisms Revealing Novel Potential Therapeutic Targets. Cancers (Basel) 2019; 11:1933. [PMID: 31817100 PMCID: PMC6966556 DOI: 10.3390/cancers11121933] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022] Open
Abstract
SerpinB3 (SB3) is a hypoxia and hypoxia-inducible factor (HIF)-2α-dependent cysteine-protease inhibitor up-regulated in hepatocellular carcinoma (HCC), released by cancer cells and able to stimulate proliferation and epithelial-to-mesenchymal-transition. Methods: In the study we employed transgenic and knock out SerpinB3 mice, liver cancer cell line, human HCC specimens, and mice receiving diethyl-nitrosamine (DEN) administration plus choline-deficient L-amino acid refined (CDAA) diet (DEN/CDAA protocol). Results: We provide detailed and mechanistic evidence that SB3 can act as a paracrine mediator able to affect the behavior of surrounding cells by differentially up-regulating, in normoxic conditions, HIF-1α and HIF-2α. SB3 acts by (i) up-regulating HIF-1α transcription, facilitating cell survival in a harsh microenvironment and promoting angiogenesis, (ii) increasing HIF-2α stabilization via direct/selective NEDDylation, promoting proliferation of liver cancer cells, and favoring HCC progression. Moreover (iii) the highest levels of NEDD8-E1 activating enzyme (NAE1) mRNA were detected in a subclass of HCC patients expressing the highest levels of HIF-2α transcripts; (iv) mice undergoing DEN/CDAA carcinogenic protocol showed a positive correlation between SB3 and HIF-2α transcripts with the highest levels of NAE1 mRNA detected in nodules expressing the highest levels of HIF-2α transcripts. Conclusions: These data outline either HIF-2α and NEDDylation as two novel putative therapeutic targets to interfere with the procarcinogenic role of SerpinB3 in the development of HCC.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Beatrice Foglia
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Gianmarco Villano
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Cristian Turato
- Veneto Institute of Oncology IOV—IRCCS, 35128 Padova, Italy;
| | - Teresa C Delgado
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Elisabetta Morello
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Fabrizio Pin
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Erica Novo
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Lucia Napione
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Torino, Italy;
- Laboratory of Vascular Oncology Candiolo Cancer Institute—FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), 10060 Candiolo, Italy;
| | - Santina Quarta
- Department of Medicine, University of Padova, 35128 Padova, Italy; (S.Q.); (P.P.)
| | - Mariagrazia Ruvoletto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Silvano Fasolato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Giacomo Zanus
- Hepatobiliary Surgery, University of Padova, 35128 Padova, Italy;
| | | | - Fernando Lopitz-Otsoa
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - David Fernández-Ramos
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Federico Bussolino
- Laboratory of Vascular Oncology Candiolo Cancer Institute—FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), 10060 Candiolo, Italy;
- Department of Oncology, University of Torino, 10125 Torino, Italy;
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Maria Luz Martínez-Chantar
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, 35128 Padova, Italy; (S.Q.); (P.P.)
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| |
Collapse
|
16
|
Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, Wang X, Lyu Y, Chen X, Liu K, Yu AM, Zuo Z, Bi H. Current trends in drug metabolism and pharmacokinetics. Acta Pharm Sin B 2019; 9:1113-1144. [PMID: 31867160 PMCID: PMC6900561 DOI: 10.1016/j.apsb.2019.10.001] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Pharmacokinetics (PK) is the study of the absorption, distribution, metabolism, and excretion (ADME) processes of a drug. Understanding PK properties is essential for drug development and precision medication. In this review we provided an overview of recent research on PK with focus on the following aspects: (1) an update on drug-metabolizing enzymes and transporters in the determination of PK, as well as advances in xenobiotic receptors and noncoding RNAs (ncRNAs) in the modulation of PK, providing new understanding of the transcriptional and posttranscriptional regulatory mechanisms that result in inter-individual variations in pharmacotherapy; (2) current status and trends in assessing drug-drug interactions, especially interactions between drugs and herbs, between drugs and therapeutic biologics, and microbiota-mediated interactions; (3) advances in understanding the effects of diseases on PK, particularly changes in metabolizing enzymes and transporters with disease progression; (4) trends in mathematical modeling including physiologically-based PK modeling and novel animal models such as CRISPR/Cas9-based animal models for DMPK studies; (5) emerging non-classical xenobiotic metabolic pathways and the involvement of novel metabolic enzymes, especially non-P450s. Existing challenges and perspectives on future directions are discussed, and may stimulate the development of new research models, technologies, and strategies towards the development of better drugs and improved clinical practice.
Collapse
Affiliation(s)
- Yuhua Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
- The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Mengbi Yang
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Xiangyu Hou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Tang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Wang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuanfeng Lyu
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ai-Ming Yu
- UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Zhong Zuo
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
17
|
Libby CJ, McConathy J, Darley-Usmar V, Hjelmeland AB. The Role of Metabolic Plasticity in Blood and Brain Stem Cell Pathophysiology. Cancer Res 2019; 80:5-16. [PMID: 31575548 DOI: 10.1158/0008-5472.can-19-1169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/04/2019] [Accepted: 09/18/2019] [Indexed: 02/06/2023]
Abstract
Our understanding of intratumoral heterogeneity in cancer continues to evolve, with current models incorporating single-cell signatures to explore cell-cell interactions and differentiation state. The transition between stem and differentiation states in nonneoplastic cells requires metabolic plasticity, and this plasticity is increasingly recognized to play a central role in cancer biology. The insights from hematopoietic and neural stem cell differentiation pathways were used to identify cancer stem cells in leukemia and gliomas. Similarly, defining metabolic heterogeneity and fuel-switching signals in nonneoplastic stem cells may also give important insights into the corresponding molecular mechanisms controlling metabolic plasticity in cancer. These advances are important, because metabolic adaptation to anticancer therapeutics is rooted in this inherent metabolic plasticity and is a therapeutic challenge to be overcome.
Collapse
Affiliation(s)
- Catherine J Libby
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jonathan McConathy
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor Darley-Usmar
- Mitochondrial Medicine Laboratory, Center for Free Radical Biology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
18
|
Wang YK, Yang XN, Liang WQ, Xiao Y, Zhao Q, Xiao XR, Gonzalez FJ, Li F. A metabolomic perspective of pazopanib-induced acute hepatotoxicity in mice. Xenobiotica 2019; 49:655-670. [PMID: 29897827 PMCID: PMC6628935 DOI: 10.1080/00498254.2018.1489167] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/09/2018] [Accepted: 06/11/2018] [Indexed: 12/27/2022]
Abstract
To elucidate the metabolism of pazopanib, a metabolomics approach was performed based on ultra-performance liquid chromatography coupled with electrospray ionization quadrupole mass spectrometry. A total of 22 pazopanib metabolites were identified in vitro and in vivo. Among these metabolites, 17 were novel, including several cysteine adducts and aldehyde derivatives. By screening using recombinant CYPs, CYP3A4 and CYP1A2 were found to be the main forms involved in the pazopanib hydroxylation. Formation of a cysteine conjugate (M3), an aldehyde derivative (M15) and two N-oxide metabolites (M18 and M20) from pazopanib could induce the oxidative stress that may be responsible in part for pazopanib-induced hepatotoxicity. Morphological observation of the liver suggested that pazopanib (300 mg/kg) could cause liver injury. The aspartate transaminase and alanine aminotransferase in serum significantly increased after pazopanib (150, 300 mg/kg) treatment; this liver injury could be partially reversed by the broad-spectrum CYP inhibitor 1-aminobenzotriazole (ABT). Metabolomics analysis revealed that pazopanib could significantly change the levels of L-carnitine, proline and lysophosphatidylcholine 18:1 in liver. Additionally, drug metabolism-related gene expression analysis revealed that hepatic Cyp2d22 and Abcb1a (P-gp) mRNAs were significantly lowered by pazopanib treatment. In conclusion, this study provides a global view of pazopanib metabolism and clues to its influence on hepatic function.
Collapse
Affiliation(s)
- Yi-Kun Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Nan Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Wei-Qing Liang
- Center for Medicinal Resources Research, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Yao Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xue-Rong Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
19
|
Abstract
Hypoxia-inducible factors (HIFs), a family of transcription factors activated by hypoxia, consist of three α-subunits (HIF1α, HIF2α and HIF3α) and one β-subunit (HIF1β), which serves as a heterodimerization partner of the HIFα subunits. HIFα subunits are stabilized from constitutive degradation by hypoxia largely through lowering the activity of the oxygen-dependent prolyl hydroxylases that hydroxylate HIFα, leading to their proteolysis. HIF1α and HIF2α are expressed in different tissues and regulate target genes involved in angiogenesis, cell proliferation and inflammation, and their expression is associated with different disease states. HIFs have been widely studied because of their involvement in cancer, and HIF2α-specific inhibitors are being investigated in clinical trials for the treatment of kidney cancer. Although cancer has been the major focus of research on HIF, evidence has emerged that this pathway has a major role in the control of metabolism and influences metabolic diseases such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease. Notably increased HIF1α and HIF2α signalling in adipose tissue and small intestine, respectively, promotes metabolic diseases in diet-induced disease models. Inhibition of HIF1α and HIF2α decreases the adverse diet-induced metabolic phenotypes, suggesting that they could be drug targets for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| |
Collapse
|
20
|
Wang YK, Xiao XR, Xu KP, Li F. Metabolic profiling of the anti-tumor drug regorafenib in mice. J Pharm Biomed Anal 2018; 159:524-535. [PMID: 30055476 DOI: 10.1016/j.jpba.2018.07.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/13/2022]
Abstract
Regorafenib is a novel tyrosine kinase inhibitor, which has been approved by the United States Food and Drug Administration for the treatment of various tumors. The purpose of the present study was to describe the metabolic map of regorafenib, and investigate its effect on liver function. Mass spectrometry-based metabolomics approach integrated with multiple mass defect filter was used to determine the metabolites of regorafenib in vitro incubation mixtures (human liver microsomes and mouse liver microsomes), serum, urine and feces samples from mice treated with 80 mg/kg regorafenib. Eleven metabolites including four novel metabolites were identified in the present investigation. As halogen substituted drug, reductive defluorination and oxidative dechlorination metabolites of regorafenib were firstly report in present study. By screening using recombinant cytochrome P450 s (CYPs), CYP3A4 was found to be the principal isoforms involved in regorafenib metabolism. The predication with a molecular docking model confirmed that regorafenib had potential to interact with the active sites of CYP3A4, CYP3A5 and CYP2D6. Serum chemistry analysis revealed no evidence of hepatic damage from regorafenib exposure. This study provided a global view of regorafenib metabolism and its potential side-effects.
Collapse
Affiliation(s)
- Yi-Kun Wang
- States Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue-Rong Xiao
- States Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Kang-Ping Xu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Fei Li
- States Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|