1
|
Tanaka N, Fukuda T, Takano R, Sasaki K, Tsuji T, Goto R, Kuribayashi T, Yamaguchi K, Niitsu Y, Ishii K, Hashimoto M, Takahashi S, Obayashi H. Discovery of DS-1093a: An oral hypoxia-inducible factor prolyl hydroxylase inhibitor for the treatment of renal anemia. Bioorg Med Chem Lett 2024; 111:129891. [PMID: 39019240 DOI: 10.1016/j.bmcl.2024.129891] [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: 04/25/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Inhibition of the hypoxia-inducible factor prolyl hydroxylase (HIF-PHD) represents a promising strategy for discovering next-generation treatments for renal anemia. We discovered DS44470011 in our previous study, which showed potent in vitro activity and in vivo efficacy based on HIF-PHD inhibition. However, DS44470011 was also found to exert genotoxic effects. By converting the biphenyl structure, which is suspected to be the cause of this genotoxicity, to a 1-phenylpiperidine structure, we were able to avoid genotoxicity and further improve the in vitro activity and in vivo efficacy. Furthermore, through the optimization of pyrimidine derivatives, we discovered DS-1093a, which has a wide safety margin with potent in vitro activity and an optimal pharmacokinetic profile. DS-1093a achieved an increase in hemoglobin levels in an adenine-induced rat model of chronic kidney disease after its continuous administration for 4 days.
Collapse
Affiliation(s)
- Naoki Tanaka
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan.
| | - Takeshi Fukuda
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Rieko Takano
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Koji Sasaki
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takashi Tsuji
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Riki Goto
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takeshi Kuribayashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kyoji Yamaguchi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yoichi Niitsu
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Ken Ishii
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masami Hashimoto
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Shinichi Takahashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hisakuni Obayashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
2
|
Gurol KC, Jursa T, Cho EJ, Fast W, Dalby KN, Smith DR, Mukhopadhyay S. PHD2 enzyme is an intracellular manganese sensor that initiates the homeostatic response against elevated manganese. Proc Natl Acad Sci U S A 2024; 121:e2402538121. [PMID: 38905240 PMCID: PMC11214094 DOI: 10.1073/pnas.2402538121] [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: 02/05/2024] [Accepted: 05/24/2024] [Indexed: 06/23/2024] Open
Abstract
Intracellular sensors detect changes in levels of essential metals to initiate homeostatic responses. But, a mammalian manganese (Mn) sensor is unknown, representing a major gap in understanding of Mn homeostasis. Using human-relevant models, we recently reported that: 1) the primary homeostatic response to elevated Mn is upregulation of hypoxia-inducible factors (HIFs), which increases expression of the Mn efflux transporter SLC30A10; and 2) elevated Mn blocks the prolyl hydroxylation of HIFs by prolyl hydroxylase domain (PHD) enzymes, which otherwise targets HIFs for degradation. Thus, the mammalian mechanism for sensing elevated Mn likely relates to PHD inhibition. Moreover, 1) Mn substitutes for a catalytic iron (Fe) in PHD structures; and 2) exchangeable cellular levels of Fe and Mn are comparable. Therefore, we hypothesized that elevated Mn directly inhibits PHD by replacing its catalytic Fe. In vitro assays using catalytically active PHD2, the primary PHD isoform, revealed that Mn inhibited, and Fe supplementation rescued, PHD2 activity. However, a mutation in PHD2 (D315E) that selectively reduced Mn binding without substantially impacting Fe binding or enzymatic activity resulted in complete insensitivity of PHD2 to Mn in vitro. Additionally, hepatic cells expressing full-length PHD2D315E were less sensitive to Mn-induced HIF activation and SLC30A10 upregulation than PHD2wild-type. These results: 1) define a fundamental Mn sensing mechanism for controlling Mn homeostasis-elevated Mn inhibits PHD2, which functions as a Mn sensor, by outcompeting its catalytic Fe, and PHD2 inhibition activates HIF signaling to up-regulate SLC30A10; and 2) identify a unique mode of metal sensing that may have wide applicability.
Collapse
Affiliation(s)
- Kerem C. Gurol
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Thomas Jursa
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Eun Jeong Cho
- College of Pharmacy, Targeted Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, TX78712
| | - Walter Fast
- Division of Chemical Biology and Drug Discovery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Kevin N. Dalby
- College of Pharmacy, Targeted Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, TX78712
- Division of Chemical Biology and Drug Discovery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
3
|
Fukuda T, Kuribayashi T, Takano R, Sasaki K, Tsuji T, Niitsu Y, Ishii K, Hashimoto M, Baba D, Ito S, Tanaka N. Discovery of DS44470011: An oral hypoxia-inducible factor prolyl hydroxylase inhibitor for the treatment of renal anemia. Bioorg Med Chem Lett 2024; 108:129799. [PMID: 38754564 DOI: 10.1016/j.bmcl.2024.129799] [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: 04/16/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Inhibition of the hypoxia-inducible factor prolyl hydroxylase (HIF-PHD) represents a promising strategy for discovering next-generation treatments for renal anemia. We identified a pyrimidine core with HIF-PHD inhibitory activity based on scaffold hopping of FG-2216 using crystal structures of HIF-PHD2 in complex with compound. By optimizing the substituents at the 2- and 6- positions of the pyrimidine core, we discovered DS44470011, which improves the effectiveness of erythropoietin (EPO) release in cells. Oral administration of DS44470011 to cynomolgus monkeys increased plasma EPO levels.
Collapse
Affiliation(s)
- Takeshi Fukuda
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan.
| | - Takeshi Kuribayashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Rieko Takano
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Koji Sasaki
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takashi Tsuji
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yoichi Niitsu
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Ken Ishii
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masami Hashimoto
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Daichi Baba
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Shuichiro Ito
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Naoki Tanaka
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
4
|
Ferens FG, Taber CC, Stuart S, Hubert M, Tarade D, Lee JE, Ohh M. Deficiency in PHD2-mediated hydroxylation of HIF2α underlies Pacak-Zhuang syndrome. Commun Biol 2024; 7:240. [PMID: 38418569 PMCID: PMC10902354 DOI: 10.1038/s42003-024-05904-4] [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: 03/30/2023] [Accepted: 02/09/2024] [Indexed: 03/01/2024] Open
Abstract
Pacak-Zhuang syndrome is caused by mutations in the EPAS1 gene, which encodes for one of the three hypoxia-inducible factor alpha (HIFα) paralogs HIF2α and is associated with defined but varied phenotypic presentations including neuroendocrine tumors and polycythemia. However, the mechanisms underlying the complex genotype-phenotype correlations remain incompletely understood. Here, we devised a quantitative method for determining the dissociation constant (Kd) of the HIF2α peptides containing disease-associated mutations and the catalytic domain of prolyl-hydroxylase (PHD2) using microscale thermophoresis (MST) and showed that neuroendocrine-associated Class 1 HIF2α mutants have distinctly higher Kd than the exclusively polycythemia-associated Class 2 HIF2α mutants. Based on the co-crystal structure of PHD2/HIF2α peptide complex at 1.8 Å resolution, we showed that the Class 1 mutated residues are localized to the critical interface between HIF2α and PHD2, adjacent to the PHD2 active catalytic site, while Class 2 mutated residues are localized to the more flexible region of HIF2α that makes less contact with PHD2. Concordantly, Class 1 mutations were found to significantly increase HIF2α-mediated transcriptional activation in cellulo compared to Class 2 counterparts. These results reveal a structural mechanism in which the strength of the interaction between HIF2α and PHD2 is at the root of the general genotype-phenotype correlations observed in Pacak-Zhuang syndrome.
Collapse
Affiliation(s)
- Fraser G Ferens
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, 661 University Avenue, Toronto, ON, M5G 1M1, Canada
| | - Cassandra C Taber
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Sarah Stuart
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, 661 University Avenue, Toronto, ON, M5G 1M1, Canada
| | - Mia Hubert
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Daniel Tarade
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Jeffrey E Lee
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Michael Ohh
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Department of Biochemistry, Faculty of Medicine, University of Toronto, 661 University Avenue, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
5
|
Chan YY, Mbenza NM, Chan MC, Leung IKH. Assays to Study Hypoxia-Inducible Factor Prolyl Hydroxylase Domain 2 (PHD2), a Key Human Oxygen Sensing Protein. Methods Mol Biol 2023; 2648:187-206. [PMID: 37039992 DOI: 10.1007/978-1-0716-3080-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Molecular oxygen is essential for all multicellular life forms. In humans, the hypoxia-inducible factor (HIF) prolyl hydroxylase domain-containing enzymes (PHDs) serve as important oxygen sensors by regulating the activity of HIF, the master regulator that mediates cellular oxygen homeostasis, in an oxygen-dependent manner. In normoxia, PHDs catalyze the prolyl hydroxylation of HIF, which leads to its degradation and prevents cellular hypoxic response to be triggered. PHDs are current inhibition targets for the potential treatments of a number of diseases. In this chapter, we discuss in vitro and cell-based methods to study the modulation of PHD2, the most important human PHD isoform in normoxia and mild hypoxia. These include the production and purification of recombinant PHD2, the use of mass spectrometry to follow PHD2-catalyzed reactions and the studies of HIF stabilization in cells by immunoblotting.
Collapse
Affiliation(s)
- Yan Ying Chan
- Department of Molecular Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Naasson M Mbenza
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mun Chiang Chan
- Department of Molecular Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Ivanhoe K H Leung
- School of Chemistry and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
6
|
Zuk A, Si Z, Loi S, Bommegowda S, Hoivik D, Danthi S, Molnar G, Csizmadia V, Rabinowitz M. Preclinical Characterization of Vadadustat (AKB-6548), an Oral Small Molecule Hypoxia-Inducible Factor Prolyl-4-Hydroxylase Inhibitor, for the Potential Treatment of Renal Anemia. J Pharmacol Exp Ther 2022; 383:11-24. [PMID: 35926869 DOI: 10.1124/jpet.122.001126] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022] Open
Abstract
Pharmacological inhibition of prolyl-4-hydroxylase domain (PHD) enzymes stabilizes hypoxia-inducible factors (HIFs), transcription factors that activate target genes that, among others, increase erythropoietin (EPO) synthesis, resulting in the production of new red blood cells (RBCs). Herein, we summarize the preclinical characteristics of the small molecule HIF prolyl-4-hydroxylase inhibitor vadadustat (AKB-6548), which is in development for the treatment of anemia in patients with chronic kidney disease (CKD). Vadadustat inhibits the enzyme activity of all three human PHD isozymes, PHD1, PHD2, and PHD3, with similar low nanomolar inhibitory constant values. PHD enzyme inhibition by vadadustat is competitive with endogenous cofactor 2-oxoglutarate and is insensitive to free iron concentration. In the human hepatocellular carcinoma cell line (Hep 3B) and human umbilical vein endothelial cells, PHD inhibition by vadadustat leads to the time- and concentration-dependent stabilization of HIF-1α and HIF-2α In Hep 3B cells, this in turn results in the synthesis and secretion of EPO; vascular endothelial growth factor is not measured at detectable levels. A single oral dose of vadadustat in rats potently increases circulating levels of EPO, and daily oral dosing for 14 days increases RBC indices in healthy rats and in the 5/6 nephrectomy model of CKD. In mice and dogs, once-daily repeat oral dosing increases hemoglobin and hematocrit. Vadadustat has a relatively short half-life in all nonclinical species evaluated and does not accumulate when administered as a single bolus dose (oral or intravenous) or upon repeat oral dosing. The pharmacological profile of vadadustat supports continued development for treatment of renal anemia. SIGNIFICANCE STATEMENT: Vadadustat (AKB-6548) is an orally bioavailable small molecule prolyl-4-hydroxylase inhibitor in development for anemia of chronic kidney disease. It is an equipotent inhibitor of the three human prolyl-4-hydroxylase domain isoforms, which activates erythropoiesis through stabilization of hypoxia-inducible factor (HIF)-1α and HIF-2α, increasing production of erythropoietin, without detectable stimulation of vascular endothelial growth factor.
Collapse
Affiliation(s)
- Anna Zuk
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Zhihai Si
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Sally Loi
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Santhosh Bommegowda
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Debie Hoivik
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Sanjay Danthi
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Gyongyi Molnar
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Vilmos Csizmadia
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| | - Michael Rabinowitz
- Department of Research and Early Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts (A.Z, Z.S., S.L, S.B., D.H., S.D., G.M., V.C., M.R)
| |
Collapse
|
7
|
Feng T, Zhao X, Gu P, Yang W, Wang C, Guo Q, Long Q, Liu Q, Cheng Y, Li J, Cheung CKY, Wu D, Kong X, Xu Y, Ye D, Hua S, Loomes K, Xu A, Hui X. Adipocyte-derived lactate is a signalling metabolite that potentiates adipose macrophage inflammation via targeting PHD2. Nat Commun 2022; 13:5208. [PMID: 36064857 PMCID: PMC9445001 DOI: 10.1038/s41467-022-32871-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Adipose tissue macrophage (ATM) inflammation is involved with meta-inflammation and pathology of metabolic complications. Here we report that in adipocytes, elevated lactate production, previously regarded as the waste product of glycolysis, serves as a danger signal to promote ATM polarization to an inflammatory state in the context of obesity. Adipocyte-selective deletion of lactate dehydrogenase A (Ldha), the enzyme converting pyruvate to lactate, protects mice from obesity-associated glucose intolerance and insulin resistance, accompanied by a lower percentage of inflammatory ATM and reduced production of pro-inflammatory cytokines such as interleukin 1β (IL-1β). Mechanistically, lactate, at its physiological concentration, fosters the activation of inflammatory macrophages by directly binding to the catalytic domain of prolyl hydroxylase domain-containing 2 (PHD2) in a competitive manner with α-ketoglutarate and stabilizes hypoxia inducible factor (HIF-1α). Lactate-induced IL-1β was abolished in PHD2-deficient macrophages. Human adipose lactate level is positively linked with local inflammatory features and insulin resistance index independent of the body mass index (BMI). Our study shows a critical function of adipocyte-derived lactate in promoting the pro-inflammatory microenvironment in adipose and identifies PHD2 as a direct sensor of lactate, which functions to connect chronic inflammation and energy metabolism.
Collapse
Affiliation(s)
- Tianshi Feng
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xuemei Zhao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ping Gu
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Wah Yang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Cunchuan Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qingyu Guo
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Qiaoyun Long
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qing Liu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ying Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin Li
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cynthia Kwan Yui Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Xinyu Kong
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Yong Xu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Dewei Ye
- Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuang Hua
- Guangdong Pharmaceutical University, Guangzhou, China
| | - Kerry Loomes
- School of Biological Sciences and Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.
- Department of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Pharmacy and Pharmacology, The University of Hong Kong, Hong Kong, China.
| | - Xiaoyan Hui
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.
- Department of Medicine, The University of Hong Kong, Hong Kong, China.
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
8
|
Zhang Y, Nsanzamahoro S, Wang CB, Wang WF, Yang JL. Screening of prolyl hydroxylase 2 inhibitors based on quantitative strategy of peptides. J Chromatogr A 2022; 1679:463411. [PMID: 35973337 DOI: 10.1016/j.chroma.2022.463411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/29/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Prolyl hydroxylase 2 (PHD2) is a key oxygen receptor regulating oxygen homeostasis in human body, and it is one of the important targets for drug research and development of hypoxia related diseases. In PHD2 enzymatic reaction, the structure of substrate (HIF-1α556-574) and product (hydroxylated HIF-1α) peptide only differ from one oxygen atom (MW>2000), which makes it a great challenge to separate them accurately and efficiently. In this work, the direct separation and detection of HIF-1α and hydroxylated HIF-1α has been firstly reported based on micellar electrokinetic chromatography (MEKC). Under optimized conditions, the intraday RSD of peak area and apparent electrophoretic mobility of hydroxylated HIF-1α were 1.87% and 0.81% respectively, and the interday RSD were 2.01% and 1.03% respectively. The LOD and LOQ of the MEKC method were 10 µM and 50 µM respectively, and the recoveries was 98.42-105.38%. Subsequently, the feasibility and accuracy of MEKC method to screen PHD2 inhibitors were confirmed by using roxadustat, and the IC50 (10.36 µM) and inhibitor type (competitive) were consistent with literature. Finally, the method was used to screen the PHD2 inhibitory activity of five traditional Chinese medicines (TCMs). The present work not only overcomes the difficulties of direct quantitative detection of hydroxylated HIF-1α, but also provides technical support for exploring and discovering new drug leads for hypoxia-related diseases from complex matrix such as TCMs.
Collapse
Affiliation(s)
- Ying Zhang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Stanislas Nsanzamahoro
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cheng-Bo Wang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Wei-Feng Wang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China.
| | - Jun-Li Yang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China.
| |
Collapse
|
9
|
Zhang Y, Zhao M, Wang CB, Wang Y, Nsanzamahoro S, Zhu LL, Wang WF, Yang JL. Screening prolyl hydroxylase domain 2 inhibitory activity of traditional Chinese medicine by CZE-UV. Electrophoresis 2022; 43:1601-1610. [PMID: 35405037 DOI: 10.1002/elps.202200028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/03/2022] [Accepted: 04/07/2022] [Indexed: 12/24/2022]
Abstract
Prolyl hydroxylase domain 2 (PHD2) is a key enzyme regulating the expression of hypoxia inducible factor (HIF). Its inhibitors can improve the expression of HIF and downstream genes, which can treat hypoxia-related diseases. Therefore, the establishment of a reliable PHD2 inhibitors screening method is of great significance for the drug development of hypoxia-related diseases. In this work, an accurate, rapid, and simple screening method for PHD2 inhibitors was introduced by capillary zone electrophoresis (CZE). In order to improve the detection sensitivity, the derivative reaction of α-ketoglutaric acid (α-OG) and 1,2-diaminobenzene (OPD) was used to enhance the UV absorption of α-OG (the substrate in the enzymatic reaction). The CZE method selected 20 mM Na2 B4 O7 buffer (pH 9.0) as the separation buffer, +25 kV as the separation voltage, 25°C as the cartridge temperature, and 210 nm as the detection wavelength. Under this condition, the analysis of a single sample can be realized within 9 min. Compared with the existing reported methods, the present work can directly screen the PHD2 inhibitory activity of traditional Chinese medicine (TCM) extracts, which is of significance for the target-purification of bioactive individual compounds from TCMs. Under the optimal conditions, the PHD2 inhibitor screening platform was successfully established, and it was found that 70% methanol/water extracts of Astragali Radix and Codonopsis pilosula had good PHD2 inhibitory activity. Furthermore, the present work provides a novel approach for screening the PHD2 inhibitory activity of TCM extracts and the discovery of anti-hypoxia bioactive compounds.
Collapse
Affiliation(s)
- Ying Zhang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Ming Zhao
- Department of Cognitive Science, Institute of Cognition and Brain Sciences, Beijing, P. R. China
| | - Cheng-Bo Wang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, P. R. China
| | - Yu Wang
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Stanislas Nsanzamahoro
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Ling-Ling Zhu
- Department of Cognitive Science, Institute of Cognition and Brain Sciences, Beijing, P. R. China
| | - Wei-Feng Wang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, P. R. China
| | - Jun-Li Yang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, P. R. China
| |
Collapse
|
10
|
Hirota K. HIF-α Prolyl Hydroxylase Inhibitors and Their Implications for Biomedicine: A Comprehensive Review. Biomedicines 2021; 9:biomedicines9050468. [PMID: 33923349 PMCID: PMC8146675 DOI: 10.3390/biomedicines9050468] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Oxygen is essential for the maintenance of the body. Living organisms have evolved systems to secure an oxygen environment to be proper. Hypoxia-inducible factor (HIF) plays an essential role in this process; it is a transcription factor that mediates erythropoietin (EPO) induction at the transcriptional level under hypoxic environment. After successful cDNA cloning in 1995, a line of studies were conducted for elucidating the molecular mechanism of HIF activation in response to hypoxia. In 2001, cDNA cloning of dioxygenases acting on prolines and asparagine residues, which play essential roles in this process, was reported. HIF-prolyl hydroxylases (PHs) are molecules that constitute the core molecular mechanism of detecting a decrease in the partial pressure of oxygen, or hypoxia, in the cells; they can be called oxygen sensors. In this review, I discuss the process of molecular cloning of HIF and HIF-PH, which explains hypoxia-induced EPO expression; the development of HIF-PH inhibitors that artificially or exogenously activate HIF by inhibiting HIF-PH; and the significance and implications of medical intervention using HIF-PH inhibitors.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
11
|
Development of a colorimetric α-ketoglutarate detection assay for prolyl hydroxylase domain (PHD) proteins. J Biol Chem 2021; 296:100397. [PMID: 33571527 PMCID: PMC7961094 DOI: 10.1016/j.jbc.2021.100397] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 11/20/2022] Open
Abstract
Since the discovery of the prolyl hydroxylases domain (PHD) proteins and their canonical hypoxia-inducible factor (HIF) substrate two decades ago, a number of in vitro hydroxylation (IVH) assays for PHD activity have been developed to measure the PHD-HIF interaction. However, most of these assays either require complex proteomics mass spectrometry methods that rely on the specific PHD-HIF interaction or require the handling of radioactive material, as seen in the most commonly used assay measuring [14C]O2 release from labeled [14C]α-ketoglutarate. Here, we report an alternative rapid, cost-effective assay in which the consumption of α-ketoglutarate is monitored by its derivatization with 2,4-dinitrophenylhydrazine (2,4-DNPH) followed by treatment with concentrated base. We extensively optimized this 2,4-DNPH α-ketoglutarate assay to maximize the signal-to-noise ratio and demonstrated that it is robust enough to obtain kinetic parameters of the well-characterized PHD2 isoform comparable with those in published literature. We further showed that it is also sensitive enough to detect and measure the IC50 values of pan-PHD inhibitors and several PHD2 inhibitors in clinical trials for chronic kidney disease (CKD)-induced anemia. Given the efficiency of this assay coupled with its multiwell format, the 2,4-DNPH α-KG assay may be adaptable to explore non-HIF substrates of PHDs and potentially to high-throughput assays.
Collapse
|
12
|
Wang RX, Henen MA, Lee JS, Vögeli B, Colgan SP. Microbiota-derived butyrate is an endogenous HIF prolyl hydroxylase inhibitor. Gut Microbes 2021; 13:1938380. [PMID: 34190032 PMCID: PMC8253137 DOI: 10.1080/19490976.2021.1938380] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota is essential for human health. Microbial supply of short-chain fatty acids (SCFAs), particularly butyrate, is a well-established contributor to gut homeostasis and disease resistance. Reaching millimolar luminal concentrations, butyrate is sequestered and utilized in the colon as the favored energy source for intestinal epithelia. Given the steep oxygen gradient across the anoxic lumen and the highly oxygenated lamina propria, the colon provides a particularly interesting environment to study oxygen sensing. Previous studies have shown that the transcription factor hypoxia-inducible factor (HIF) is stabilized in healthy colonic epithelia. Here we show that butyrate directly inhibits HIF prolyl hydroxylases (PHDs) to stabilize HIF. We find that butyrate stabilizes HIF in vitro despite eliminating β-oxidation and resultant oxygen consumption. Using recombinant PHD protein in combination with nuclear magnetic resonance and enzymatic biochemical assays, we identify butyrate to bind and function as a unique, noncompetitive inhibitor of PHDs relative to other SCFAs. Butyrate inhibited PHD with a noncompetitive Ki of 5.3 ± 0.5 mM, a physiologically relevant concentration. We also confirm that microbiota-derived butyrate is necessary to stabilize HIF in mice colonic tissue through antibiotic-induced butyrate depletion and reconstitution experiments. Our results suggest that the co-evolution of mammals and mutualistic microbiota has selected for butyrate to impact a critical gene regulation pathway that can be extended beyond the mammalian gut. As PHDs are a major target for drug development in the stabilization of HIF, butyrate holds great potential as a well-tolerated endogenous inhibitor with far-reaching therapeutic impact.
Collapse
Affiliation(s)
- Ruth X. Wang
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- School of Medicine, Medical Scientist Training Program, University of Colorado, Aurora, CO, USA
| | - Morkos A. Henen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmaceutical Organic Chemistry, Mansoura University, Mansoura, Egypt
| | - J. Scott Lee
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Beat Vögeli
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sean P. Colgan
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
13
|
Mitochondrial MUL1 E3 ubiquitin ligase regulates Hypoxia Inducible Factor (HIF-1α) and metabolic reprogramming by modulating the UBXN7 cofactor protein. Sci Rep 2020; 10:1609. [PMID: 32005965 PMCID: PMC6994496 DOI: 10.1038/s41598-020-58484-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/18/2019] [Indexed: 11/15/2022] Open
Abstract
MUL1 is a multifunctional E3 ubiquitin ligase anchored in the outer mitochondrial membrane with its RING finger domain facing the cytoplasm. MUL1 participates in various biological pathways involved in apoptosis, mitochondrial dynamics, and innate immune response. The unique topology of MUL1 enables it to “sense” mitochondrial stress in the intermembrane mitochondrial space and convey these signals through the ubiquitination of specific cytoplasmic substrates. We have identified UBXN7, the cofactor protein of the CRL2VHL ligase complex, as a specific substrate of MUL1 ligase. CRL2VHL ligase complex regulates HIF-1α protein levels under aerobic (normoxia) or anaerobic (hypoxia) conditions. Inactivation of MUL1 ligase leads to accumulation of UBXN7, with concomitant increase in HIF-1α protein levels, reduction in oxidative phosphorylation, and increased glycolysis. We describe a novel pathway that originates in the mitochondria and operates upstream of the CRL2VHL ligase complex. Furthermore, we delineate the mechanism by which the mitochondria, through MUL1 ligase, can inhibit the CRL2VHL complex leading to high HIF-1α protein levels and a metabolic shift to glycolysis under normoxic conditions.
Collapse
|
14
|
Seim GL, Britt EC, John SV, Yeo FJ, Johnson AR, Eisenstein RS, Pagliarini DJ, Fan J. Two-stage metabolic remodelling in macrophages in response to lipopolysaccharide and interferon-γ stimulation. Nat Metab 2019; 1:731-742. [PMID: 32259027 PMCID: PMC7108803 DOI: 10.1038/s42255-019-0083-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 06/07/2019] [Indexed: 12/13/2022]
Abstract
In response to signals associated with infection or tissue damage, macrophages undergo a series of dynamic phenotypic changes. Here we show that during the response to LPS and interferon-γ stimulation, metabolic reprogramming in macrophages is also highly dynamic. Specifically, the TCA cycle undergoes a two-stage remodeling: the early stage is characterized by a transient accumulation of intermediates including succinate and itaconate, while the late stage is marked by the subsidence of these metabolites. The metabolic transition into the late stage is largely driven by the inhibition of pyruvate dehydrogenase complex (PDHC) and oxoglutarate dehydrogenase complex (OGDC), which is controlled by the dynamic changes in lipoylation state of both PDHC and OGDC E2 subunits and phosphorylation of PDHC E1 subunit. This dynamic metabolic reprogramming results in a transient metabolic state that strongly favors HIF-1α stabilization during the early stage, which subsides by the late stage; consistently, HIF-1α levels follow this trend. This study elucidates a dynamic and mechanistic picture of metabolic reprogramming in LPS and interferon-γ stimulated macrophages, and provides insights into how changing metabolism can regulate the functional transitions in macrophages over a course of immune response.
Collapse
Affiliation(s)
- Gretchen L Seim
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Emily C Britt
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Steven V John
- Morgridge Institute for Research, Madison, WI
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | | | | | - Richard S Eisenstein
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- Morgridge Institute for Research, Madison, WI
| | - David J Pagliarini
- Morgridge Institute for Research, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, Madison, WI
- Correspondence to
| |
Collapse
|
15
|
The multifaceted contribution of α-ketoglutarate to tumor progression: An opportunity to exploit? Semin Cell Dev Biol 2019; 98:26-33. [PMID: 31175937 DOI: 10.1016/j.semcdb.2019.05.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 01/25/2023]
Abstract
The thriving field that constitutes cancer metabolism has unveiled some groundbreaking facts over the past two decades, at the heart of which is the TCA cycle and its intermediates. As such and besides its metabolic role, α-ketoglutarate was shown to withstand a wide range of physiological reactions from protection against oxidative stress, collagen and bone maintenance to development and immunity. Most importantly, it constitutes the rate-limiting substrate of 2-oxoglutarate-dependent dioxygenases family enzymes, which are involved in hypoxia sensing and in the shaping of cellular epigenetic landscape, two major drivers of oncogenic transformation. Based on literature reports, we hereby review the benefits of this metabolite as a possible novel adjuvant therapeutic opportunity to target tumor progression. This article is part of the special issue "Mitochondrial metabolic alterations in cancer cells and related therapeutic targets".
Collapse
|
16
|
Manresa MC, Smith L, Casals‐Diaz L, Fagundes RR, Brown E, Radhakrishnan P, Murphy SJ, Crifo B, Strowitzki MJ, Halligan DN, van den Bogaard EH, Niehues H, Schneider M, Taylor CT, Steinhoff M. Pharmacologic inhibition of hypoxia-inducible factor (HIF)-hydroxylases ameliorates allergic contact dermatitis. Allergy 2019; 74:753-766. [PMID: 30394557 DOI: 10.1111/all.13655] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND When an immune cell migrates from the bloodstream to a site of chronic inflammation, it experiences a profound decrease in microenvironmental oxygen levels leading to a state of cellular hypoxia. The hypoxia-inducible factor-1α (HIF-1α) promotes an adaptive transcriptional response to hypoxia and as such is a major regulator of immune cell survival and function. HIF hydroxylases are the family of oxygen-sensing enzymes primarily responsible for conferring oxygen dependence upon the HIF pathway. METHODS Using a mouse model of allergic contact dermatitis (ACD), we tested the effects of treatment with the pharmacologic hydroxylase inhibitor DMOG, which mimics hypoxia, on disease development. RESULTS Re-exposure of sensitized mice to 2,4-dinitrofluorobenzene (DNFB) elicited inflammation, edema, chemokine synthesis (including CXCL1 and CCL5) and the recruitment of neutrophils and eosinophils. Intraperitoneal or topical application of the pharmacologic hydroxylase inhibitors dymethyloxalylglycine (DMOG) or JNJ1935 attenuated this inflammatory response. Reduced inflammation was associated with diminished recruitment of neutrophils and eosinophils but not lymphocytes. Finally, hydroxylase inhibition reduced cytokine-induced chemokine production in cultured primary keratinocytes through attenuation of the JNK pathway. CONCLUSION These data demonstrate that hydroxylase inhibition attenuates the recruitment of neutrophils to inflamed skin through reduction of chemokine production and increased neutrophilic apoptosis. Thus, pharmacologic inhibition of HIF hydroxylases may be an effective new therapeutic approach in allergic skin inflammation.
Collapse
Affiliation(s)
- Mario C. Manresa
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Leila Smith
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Laura Casals‐Diaz
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Raphael R. Fagundes
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Eric Brown
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery University of Heidelberg Heidelberg Germany
| | - Stephen J. Murphy
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Bianca Crifo
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Moritz J. Strowitzki
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Doug N. Halligan
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Ellen H. van den Bogaard
- Department of Dermatology Radboud University Medical Center Radboud Institute for Molecular Life Sciences Nijmegen The Netherlands
| | - Hanna Niehues
- Department of Dermatology Radboud University Medical Center Radboud Institute for Molecular Life Sciences Nijmegen The Netherlands
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery University of Heidelberg Heidelberg Germany
| | - Cormac T. Taylor
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Conway Institute of Biomedical and Biomolecular Research School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Systems Biology Ireland School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
| | - Martin Steinhoff
- UCD Charles Institute of Dermatology School of Medicine and Medical Science University College Dublin Belfield, Dublin Ireland
- Department of Dermatology & Venereology Translational Research Institute Hamad Medical Corporation Weill Cornell University‐Qatar and Qatar University Doha Qatar
| |
Collapse
|
17
|
Yeldag G, Rice A, Del Río Hernández A. Chemoresistance and the Self-Maintaining Tumor Microenvironment. Cancers (Basel) 2018; 10:E471. [PMID: 30487436 PMCID: PMC6315745 DOI: 10.3390/cancers10120471] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
The progression of cancer is associated with alterations in the tumor microenvironment, including changes in extracellular matrix (ECM) composition, matrix rigidity, hypervascularization, hypoxia, and paracrine factors. One key malignant phenotype of cancer cells is their ability to resist chemotherapeutics, and elements of the ECM can promote chemoresistance in cancer cells through a variety of signaling pathways, inducing changes in gene expression and protein activity that allow resistance. Furthermore, the ECM is maintained as an environment that facilitates chemoresistance, since its constitution modulates the phenotype of cancer-associated cells, which themselves affect the microenvironment. In this review, we discuss how the properties of the tumor microenvironment promote chemoresistance in cancer cells, and the interplay between these external stimuli. We focus on both the response of cancer cells to the external environment, as well as the maintenance of the external environment, and how a chemoresistant phenotype emerges from the complex signaling network present.
Collapse
Affiliation(s)
- Gulcen Yeldag
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| | - Armando Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
18
|
Tarade D, Robinson CM, Lee JE, Ohh M. HIF-2α-pVHL complex reveals broad genotype-phenotype correlations in HIF-2α-driven disease. Nat Commun 2018; 9:3359. [PMID: 30135421 PMCID: PMC6105673 DOI: 10.1038/s41467-018-05554-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022] Open
Abstract
It is definitively established that mutations in transcription factor HIF-2α are causative of both neuroendocrine tumors (class 1 disease) and polycythemia (class 2 disease). However, the molecular mechanism that underlies this emergent genotype–phenotype relationship has remained unclear. Here, we report the structure of HIF-2α peptide bound to pVHL-elongin B-elongin C (VBC) heterotrimeric complex, which shows topographical demarcation of class 1 and 2 mutations affecting residues predicted, and demonstrated via biophysical analyses, to differentially impact HIF-2α-pVHL interaction interface stability. Concordantly, biochemical experiments showed that class 1 mutations disrupt pVHL affinity to HIF-2α more adversely than class 2 mutations directly or indirectly via impeding PHD2-mediated hydroxylation. These findings suggest that neuroendocrine tumor pathogenesis requires a higher HIF-2α dose than polycythemia, which requires only a mild increase in HIF-2α activity. These biophysical data reveal a structural basis that underlies, and can be used to predict de novo, broad genotype-phenotype correlations in HIF-2α-driven disease. Hypoxia inducible factor (HIF)-2α transcription factor is mutated in polycythemia and various neuroendocrine tumors. Here the authors present the crystal structure of a HIF-2α peptide bound to the pVHL-elongin B-elongin C (VBC) heterotrimeric complex and propose a classification scheme for HIF-2α mutations that helps to predict disease phenotype outcome.
Collapse
Affiliation(s)
- Daniel Tarade
- Department of Laboratory Medicine & Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Claire M Robinson
- Department of Laboratory Medicine & Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Department of Biochemistry, University of Toronto, 661 University Avenue, Toronto, ON, M5G 1M1, Canada
| | - Jeffrey E Lee
- Department of Laboratory Medicine & Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Michael Ohh
- Department of Laboratory Medicine & Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Department of Biochemistry, University of Toronto, 661 University Avenue, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
19
|
Discovery of novel 2-[(4-hydroxy-6-oxo-2,3-dihydro-1H-pyridine-5-carbonyl)amino]acetic acid derivatives as HIF prolyl hydroxylase inhibitors for treatment of renal anemia. Bioorg Med Chem Lett 2018; 28:1725-1730. [DOI: 10.1016/j.bmcl.2018.04.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/14/2018] [Indexed: 12/20/2022]
|
20
|
Smirnova NA, Osipyants AI, Khristichenko AY, Hushpulian DM, Nikulin SV, Chubar TA, Zakhariants AA, Tishkov VI, Gazaryan IG, Poloznikov AA. HIF2 ODD-luciferase reporter: the most sensitive assay for HIF prolyl hydroxylase inhibitors. Russ Chem Bull 2018. [DOI: 10.1007/s11172-018-2051-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Abstract
Kinetic analyses of HIF prolyl 4-hydroxylases (HIF-P4Hs) allow determination of substrate, cosubstrate and cofactor requirements, analysis of the reaction rate, and inhibitory properties of the isoenzymes in vitro. Here we describe an assay measuring the substrate hydroxylation-coupled decarboxylation of radioactive 2-oxoglutarate to radioactive carbon dioxide as a fast, efficient, and diverse method to analyze the enzyme kinetics of HIF-P4Hs.
Collapse
Affiliation(s)
- Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, Biocenter Oulu, University of Oulu, Oulu, Finland.
| | - Johanna Myllyharju
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
22
|
Ma M, Hua S, Li G, Wang S, Cheng X, He S, Wu P, Chen X. Prolyl hydroxylase domain protein 3 and asparaginyl hydroxylase factor inhibiting HIF-1 levels are predictive of tumoral behavior and prognosis in hepatocellular carcinoma. Oncotarget 2017; 8:12983-13002. [PMID: 28099905 PMCID: PMC5355071 DOI: 10.18632/oncotarget.14677] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/09/2017] [Indexed: 01/22/2023] Open
Abstract
Hypoxia-inducible factors (HIFs) are key regulators in oxygen homeostasis. Their stabilization and activity are regulated by prolyl hydroxylase domain (PHD)-1, -2, -3 and factor inhibiting HIF (FIH). This study investigated the relation between these oxygen sensors and the clinical behaviors and prognosis of hepatocellular carcinoma (HCC). Tissue microarray and RT-PCR analysis of tumor tissues and adjacent non-tumor liver tissues revealed that mRNA and protein levels of both PHD3 and FIH were lower within tumors. The lower expression of PHD3 in tumor was associated with larger tumor size, incomplete tumor encapsulation, vascular invasion and higher Ki-67 LI (p < 0.05). The lower expression of FIH in tumor was associated with incomplete tumor encapsulation, vascular invasion, as well as higher TNM stage, BCLC stage, microvascular density and Ki-67 LI (p < 0.05). Patients with reduced expression of PHD3 or FIH had markedly shorter disease-free survival (DFS), lower overall survival (OS), or higher recurrence (p < 0.05), especially early recurrence. Patients with simultaneously reduced expression of PHD3 and FIH exhibited the least chance of forming tumor encapsulation, highest TNM stage (p < 0.0083), lowest OS and highest recurrence rate (p < 0.05). Multivariate analysis indicated that a lower expression of FIH independently predicted a poor prognosis in HCC. These findings indicate that downregulation of PHD3 and FIH in HCC is associated with more aggressive tumor behavior and a poor prognosis. PHD3 and FIH may be potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Mingyang Ma
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan 430030, China
| | - Shuyao Hua
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gang Li
- Department of Surgery, Liyuan Hospital, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Sumei Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Cheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Songqing He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.,Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China.,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin 541001, China
| | - Ping Wu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan 430030, China
| |
Collapse
|
23
|
Chromatin remodeling system p300-HDAC2-Sin3A is involved in Arginine Starvation-Induced HIF-1α Degradation at the ASS1 promoter for ASS1 Derepression. Sci Rep 2017; 7:10814. [PMID: 28883660 PMCID: PMC5589935 DOI: 10.1038/s41598-017-11445-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/17/2022] Open
Abstract
Argininosuccinate synthetase 1 (ASS1) is the key enzyme that controls biosynthesis of arginine (Arg). ASS1 is silenced in many human malignancies therefore, these tumors require extracellular Arg for growth. The Arg-degrading recombinant protein, pegylated arginine deiminase (ADI-PEG20), has been in clinical trials for targeting Arg auxotrophic tumors by Arg starvation therapy. Resistance to Arg starvation is often developed through reactivation of ASS1 expression. We previously demonstrated that ASS1 silencing is controlled by HIF-1α and Arg starvation-reactivated ASS1 is associated with HIF-1α downregulation. However, mechanisms underlying ASS1 repression and HIF-1α turnover are not known. Here, we demonstrate that interplay of p300-HDAC2-Sin3A in the chromatin remodeling system is involved in HIF-1α degradation at the ASS1 promoter. The histone acetyltransferase p300 is normally associated with the ASS1 promoter to maintain acetylated H3K14ac and H3K27ac for ASS1 silencing. Arg starvation induces p300 dissociation, allowing histone HDAC2 and cofactor Sin3A to deacetylate these histones at the ASS1 promoter, thereby facilitating HIF-1α-proteasomal complex, driven by PHD2, to degrade HIF-1α in situ. Arg starvation induces PHD2 and HDAC2 interaction which is sensitive to antioxidants. This is the first report describing epigenetic regulation of chromosomal HIF-1α turnover in gene activation that bears important implication in cancer therapy.
Collapse
|
24
|
Manresa MC, Taylor CT. Hypoxia Inducible Factor (HIF) Hydroxylases as Regulators of Intestinal Epithelial Barrier Function. Cell Mol Gastroenterol Hepatol 2017; 3:303-315. [PMID: 28462372 PMCID: PMC5404106 DOI: 10.1016/j.jcmgh.2017.02.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
Abstract
Human health is dependent on the ability of the body to extract nutrients, fluids, and oxygen from the external environment while at the same time maintaining a state of internal sterility. Therefore, the cell layers that cover the surface areas of the body such as the lung, skin, and gastrointestinal mucosa provide vital semipermeable barriers that allow the transport of essential nutrients, fluid, and waste products, while at the same time keeping the internal compartments free of microbial organisms. These epithelial surfaces are highly specialized and differ in their anatomic structure depending on their location to provide appropriate and effective site-specific barrier function. Given this important role, it is not surprising that significant disease often is associated with alterations in epithelial barrier function. Examples of such diseases include inflammatory bowel disease, chronic obstructive pulmonary disease, and atopic dermatitis. These chronic inflammatory disorders often are characterized by diminished tissue oxygen levels (hypoxia). Hypoxia triggers an adaptive transcriptional response governed by hypoxia-inducible factors (HIFs), which are repressed by a family of oxygen-sensing HIF hydroxylases. Here, we review recent evidence suggesting that pharmacologic hydroxylase inhibition may be of therapeutic benefit in inflammatory bowel disease through the promotion of intestinal epithelial barrier function through both HIF-dependent and HIF-independent mechanisms.
Collapse
Key Words
- CD, Crohn’s disease
- DMOG, dimethyloxalylglycine
- DSS, dextran sodium sulfate
- Epithelial Barrier
- FIH, factor inhibiting hypoxia-inducible factor
- HIF, hypoxia-inducible factor
- Hypoxia
- Hypoxia-Inducible Factor (HIF) Hydroxylases
- IBD, inflammatory bowel disease
- IL, interleukin
- Inflammatory Bowel Disease
- NF-κB, nuclear factor-κB
- PHD, hypoxia-inducible factor–prolyl hydroxylases
- TFF, trefoil factor
- TJ, tight junction
- TLR, Toll-like receptor
- TNF-α, tumor necrosis factor α
- UC, ulcerative colitis
- ZO, zonula occludens
Collapse
Affiliation(s)
- Mario C. Manresa
- Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin, Ireland
- Charles Institute of Dermatology, Belfield, Dublin, Ireland
| | - Cormac T. Taylor
- Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin, Ireland
- Charles Institute of Dermatology, Belfield, Dublin, Ireland
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
25
|
Modulation of TRP Channel Activity by Hydroxylation and Its Therapeutic Potential. Pharmaceuticals (Basel) 2017; 10:ph10020035. [PMID: 28346371 PMCID: PMC5490392 DOI: 10.3390/ph10020035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/15/2017] [Accepted: 03/24/2017] [Indexed: 12/15/2022] Open
Abstract
Two transient receptor potential (TRP) channels—TRPA1 and TRPV3—are post-translationally hydroxylated, resulting in oxygen-dependent regulation of channel activity. The enzymes responsible are the HIF prolyl hydroxylases (PHDs) and the asparaginyl hydroxylase factor inhibiting HIF (FIH). The PHDs and FIH are well characterized for their hydroxylation of the hypoxic inducible transcription factors (HIFs), mediating their hypoxic regulation. Consequently, these hydroxylases are currently being targeted therapeutically to modulate HIF activity in anemia, inflammation, and ischemic disease. Modulating the HIFs by targeting these hydroxylases may result in both desirable and undesirable effects on TRP channel activity, depending on the physiological context. For the best outcomes, these hydroxylases could be therapeutically targeted in pathologies where activation of both the HIFs and the relevant TRP channels are predicted to independently achieve positive outcomes, such as wound healing and obesity.
Collapse
|
26
|
Zhang H, Shang P, Zhang Y, Zhang Y, Tian X, Zhang H. Angiogenesis in chicken chorioallantoic membrane under hypoxic incubations. ANIM BIOL 2017. [DOI: 10.1163/15707563-00002540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Under hypoxic conditions, angiogenesis in the chorioallantoic membrane exhibits a sensitive response depending on the developmental timing and intensity and duration of the hypoxia. Furthermore, the effects of hypoxia on vascularization in the chorioallantoic membrane are controversial. In this study, we used microscopy to determine the vascular density index of chicken embryonic chorioallantoic membrane and quantitative PCR to examine the expression of the HIF-1α and VEGFA genes. Two levels of hypoxia (15% and 13% O2) and three durations (days 0–10, 11–18, and 0–18) were applied. The results showed that 13% O2 incubation restrained angiogenesis in the chorioallantoic membrane at an early stage of embryonic development as seen on day 6, but incubation under 13% or 15% O2 efficiently stimulated vascularization of the chorioallantoic membrane as seen on days 16 and 18. Notably, 13% O2 incubation caused visible curling of the blood vessels in the chorioallantoic membrane on day 18. The increased vascular density index under hypoxia was accompanied by an increase in the expression of VEGFA and HIF-1α. Curled blood vessels were observed on day 18 under 13% O2 incubation; however, the curling was not a result of VEGFA overexpression. Thus, the present study helps in elucidating vascularization of the chorioallantoic membrane under hypoxic conditions.
Collapse
Affiliation(s)
- Hongliang Zhang
- 1National Engineering Laboratory for Animal Breeding/Laboratory of Animal Genetic Resource and Molecular Breeding, China Agricultural University, No. 2 Yuanmingyuan West Rd., Beijing, China
| | - Peng Shang
- 1National Engineering Laboratory for Animal Breeding/Laboratory of Animal Genetic Resource and Molecular Breeding, China Agricultural University, No. 2 Yuanmingyuan West Rd., Beijing, China
- 2College of Animal Science, Tibet Agriculture and Animal Husbandry College, Linzhi, Tibet, China
| | - Yawen Zhang
- 1National Engineering Laboratory for Animal Breeding/Laboratory of Animal Genetic Resource and Molecular Breeding, China Agricultural University, No. 2 Yuanmingyuan West Rd., Beijing, China
| | - Ying Zhang
- 1National Engineering Laboratory for Animal Breeding/Laboratory of Animal Genetic Resource and Molecular Breeding, China Agricultural University, No. 2 Yuanmingyuan West Rd., Beijing, China
| | - Xiaolong Tian
- 1National Engineering Laboratory for Animal Breeding/Laboratory of Animal Genetic Resource and Molecular Breeding, China Agricultural University, No. 2 Yuanmingyuan West Rd., Beijing, China
| | - Hao Zhang
- 1National Engineering Laboratory for Animal Breeding/Laboratory of Animal Genetic Resource and Molecular Breeding, China Agricultural University, No. 2 Yuanmingyuan West Rd., Beijing, China
| |
Collapse
|
27
|
Youssef S, Ren W, Ai HW. A Genetically Encoded FRET Sensor for Hypoxia and Prolyl Hydroxylases. ACS Chem Biol 2016; 11:2492-8. [PMID: 27385075 DOI: 10.1021/acschembio.6b00330] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Oxygen is vital for all aerobic life forms. Oxygen-dependent hydroxylation of hypoxia-inducible factor (HIF)-1α by prolyl hydroxylase domain enzymes (PHDs) is an important step for controlling the expression of oxygen-regulated genes in metazoan species, thereby constituting a molecular mechanism for oxygen sensing and response. Herein, we report a genetically encoded dual-emission ratiometric fluorescent sensor, ProCY, which responds to PHD activities in vitro and in live cells. We demonstrated that ProCY could monitor hypoxia in mammalian cells. By targeting this novel genetically encoded biosensor to the cell nucleus and cytosol, we determined that, under normoxic conditions, the HIF-prolyl hydroxylase activity was mainly confined to the cytosol of HEK 293T cells. The results collectively suggest broad applications of ProCY on the evaluation of hypoxia and PHD activities and understanding of pathways for the control of hypoxic responses.
Collapse
Affiliation(s)
- Suzan Youssef
- Department of Chemistry, University of California, Riverside, 501 Big Springs Road, Riverside, California 92521, United States
| | - Wei Ren
- Department of Chemistry, University of California, Riverside, 501 Big Springs Road, Riverside, California 92521, United States
| | - Hui-wang Ai
- Department of Chemistry, University of California, Riverside, 501 Big Springs Road, Riverside, California 92521, United States
| |
Collapse
|
28
|
Vasta JD, Raines RT. Human Collagen Prolyl 4-Hydroxylase Is Activated by Ligands for Its Iron Center. Biochemistry 2016; 55:3224-33. [PMID: 27183028 DOI: 10.1021/acs.biochem.6b00251] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Collagen is the most abundant protein in animals. The posttranslational hydroxylation of proline residues in collagen contributes greatly to its conformational stability. Deficient hydroxylation is associated with a variety of disease states, including scurvy. The hydroxylation of proline residues in collagen is catalyzed by an Fe(II)- and α-ketoglutarate-dependent dioxygenase, collagen prolyl 4-hydroxylase (CP4H). CP4H has long been known to suffer oxidative inactivation during catalysis, and the cofactor ascorbate (vitamin C) is required to reactivate the enzyme by reducing its iron center from Fe(III) to Fe(II). Herein, we report on the discovery of the first synthetic activators of CP4H. Specifically, we find that 2,2'-bipyridine-4-carboxylate and 2,2'-bipyridine-5-carboxylate serve as ligands for the iron center in human CP4H that enhance the rate of ascorbate-dependent reactivation. This new mode of CP4H activation is available to other biheteroaryl compounds but does not necessarily extend to other prolyl 4-hydroxylases. As collagen is weakened in many indications, analogous activators of CP4H could have therapeutic benefits.
Collapse
Affiliation(s)
- James D Vasta
- Department of Biochemistry and ‡Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Ronald T Raines
- Department of Biochemistry and ‡Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| |
Collapse
|
29
|
Hadi-Alijanvand H, Proctor EA, Ding F, Dokholyan NV, Moosavi-Movahedi AA. A hidden aggregation-prone structure in the heart of hypoxia inducible factor prolyl hydroxylase. Proteins 2016; 84:611-23. [DOI: 10.1002/prot.25011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Hamid Hadi-Alijanvand
- Department of Biological Sciences; Institute for Advanced Studies in Basic Sciences (IASBS); Zanjan Iran
- Institute of Biochemistry and Biophysics (IBB), University of Tehran; Tehran Iran
| | - Elizabeth A. Proctor
- Department of Biological Engineering; Massachusetts Institute of Technology; Cambridge Massachusetts 02139
| | - Feng Ding
- Department of Biochemistry and Biophysics; University of North Carolina at Chapel Hill, School of Medicine; Chapel Hill North Carolina 27599
- Department of Physics and Astronomy; Clemson University; Clemson South Carolina 29634
| | - Nikolay V. Dokholyan
- Department of Biochemistry and Biophysics; University of North Carolina at Chapel Hill, School of Medicine; Chapel Hill North Carolina 27599
- Curriculum in Bioinformatics and Computational Biology; University of North Carolina at Chapel Hill, School of Medicine; Chapel Hill North Carolina 27599
- Program in Molecular and Cellular Biophysics; University of North Carolina at Chapel Hill, School of Medicine; Chapel Hill North Carolina 27599
| | - Ali A. Moosavi-Movahedi
- Institute of Biochemistry and Biophysics (IBB), University of Tehran; Tehran Iran
- Center of Excellence in Biothermodynamics, Institute of Biochemistry and Biophysics (IBB), University of Tehran; Tehran Iran
| |
Collapse
|
30
|
Pharmacological targeting of the HIF hydroxylases--A new field in medicine development. Mol Aspects Med 2016; 47-48:54-75. [PMID: 26791432 DOI: 10.1016/j.mam.2016.01.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/11/2015] [Accepted: 01/04/2016] [Indexed: 12/13/2022]
Abstract
In human cells oxygen levels are 'sensed' by a set of ferrous iron and 2-oxoglutarate dependent dioxygenases. These enzymes regulate a broad range of cellular and systemic responses to hypoxia by catalysing the post-translational hydroxylation of specific residues in the alpha subunits of hypoxia inducible factor (HIF) transcriptional complexes. The HIF hydroxylases are now the subject of pharmaceutical targeting by small molecule inhibitors that aim to activate or augment the endogenous HIF transcriptional response for the treatment of anaemia and other hypoxic human diseases. Here we consider the rationale for this therapeutic strategy from the biochemical, biological and medical perspectives. We outline structural and mechanistic considerations that are relevant to the design of HIF hydroxylase inhibitors, including likely determinants of specificity, and review published reports on their activity in pre-clinical models and clinical trials.
Collapse
|
31
|
Yalu R, Oyesiji AE, Eisenberg I, Imbar T, Meidan R. HIF1A-dependent increase in endothelin 2 levels in granulosa cells: role of hypoxia, LH/cAMP, and reactive oxygen species. Reproduction 2015; 149:11-20. [PMID: 25433027 DOI: 10.1530/rep-14-0409] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxia-inducible factor 1 alpha (HIF1A) and endothelin 2 (EDN2) are transiently expressed during the same time window in the developing corpus luteum (CL). In this study, we sought to investigate the involvement of LH/cAMP, reactive oxygen species (ROS), and a hypoxia-mimetic compound (CoCl2) on HIF1A expression and how it affected EDN2 levels, using transformed human granulosa cells (thGCs) and primary bovine granulosa cells (GCs). CoCl2 elevated HIF1A protein levels in thGCs in a dose-dependent manner. Forskolin alone had no significant effect; however, forskolin and CoCl2 together further induced HIF1A protein and EDN2 mRNA expression in thGCs. Similarly, in primary GCs, LH with CoCl2 synergistically augmented HIF1A protein levels, which resulted in higher expression of EDN2 and another well-known hypoxia-inducible gene, VEGF (VEGFA). Importantly, LH alone elevated HIF1A mRNA but not its protein. The successful knockdown of HIF1A in thGCs using siRNA abolished hypoxia-induced EDN2 and also the additive effect of forskolin and CoCl2. We then examined the roles of ROS in thGCs: hydrogen peroxide (20 and 50 μM) elevated HIF1A protein as well as the expression of EDN2, implying that induction of HIF1A protein levels is sufficient to stimulate the expression of EDN2 (and VEGF) in normoxia. A broad-range ROS scavenger, butylated hydroxyanisole, inhibited CoCl2-induced HIF1A protein with a concomitant reduction in the mRNA expression of EDN2 and VEGF in thGCs. The results obtained in this study suggest that HIF1A, induced by various stimuli, is an essential mediator of EDN2 mRNA expression. The results may also explain the rise in the levels of HIF1A-dependent genes (EDN2 and VEGF) in the developing CL.
Collapse
Affiliation(s)
- Ronit Yalu
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Adepeju Esther Oyesiji
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Iris Eisenberg
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Tal Imbar
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Rina Meidan
- Department of Animal SciencesThe Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Herzl Street, Rehovot 76100, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| |
Collapse
|
32
|
Vasta JD, Raines RT. Selective inhibition of prolyl 4-hydroxylases by bipyridinedicarboxylates. Bioorg Med Chem 2015; 23:3081-90. [PMID: 26022078 DOI: 10.1016/j.bmc.2015.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 04/27/2015] [Accepted: 05/03/2015] [Indexed: 10/23/2022]
Abstract
Collagen is the most abundant protein in animals. A variety of indications are associated with the overproduction of collagen, including fibrotic diseases and cancer metastasis. The stability of collagen relies on the posttranslational modification of proline residues to form (2S,4R)-4-hydroxyproline. This modification is catalyzed by collagen prolyl 4-hydroxylases (CP4Hs), which are Fe(II)- and α-ketoglutarate (AKG)-dependent dioxygenases located in the lumen of the endoplasmic reticulum. Human CP4Hs are validated targets for treatment of both fibrotic diseases and metastatic breast cancer. Herein, we report on 2,2'-bipyridinedicarboxylates as inhibitors of a human CP4H. Although most 2,2'-bipyridinedicarboxylates are capable of inhibition via iron sequestration, the 4,5'- and 5,5'-dicarboxylates were found to be potent competitive inhibitors of CP4H, and the 5,5'-dicarboxylate was selective in its inhibitory activity. Our findings clarify a strategy for developing CP4H inhibitors of clinical utility.
Collapse
Affiliation(s)
- James D Vasta
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Ronald T Raines
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, United States; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States.
| |
Collapse
|
33
|
Gardie B, Percy MJ, Hoogewijs D, Chowdhury R, Bento C, Arsenault PR, Richard S, Almeida H, Ewing J, Lambert F, McMullin MF, Schofield CJ, Lee FS. The role of PHD2 mutations in the pathogenesis of erythrocytosis. HYPOXIA 2014; 2:71-90. [PMID: 27774468 PMCID: PMC5045058 DOI: 10.2147/hp.s54455] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The transcription of the erythropoietin (EPO) gene is tightly regulated by the hypoxia response pathway to maintain oxygen homeostasis. Elevations in serum EPO level may be reflected in an augmentation in the red cell mass, thereby causing erythrocytosis. Studies on erythrocytosis have provided insights into the function of the oxygen-sensing pathway and the critical proteins involved in the regulation of EPO transcription. The α subunits of the hypoxia-inducible transcription factor are hydroxylated by three prolyl hydroxylase domain (PHD) enzymes, which belong to the iron and 2-oxoglutarate-dependent oxygenase superfamily. Sequence analysis of the genes encoding the PHDs in patients with erythrocytosis has revealed heterozygous germline mutations only occurring in Egl nine homolog 1 (EGLN1, also known as PHD2), the gene that encodes PHD2. To date, 24 different EGLN1 mutations comprising missense, frameshift, and nonsense mutations have been described. The phenotypes associated with the patients carrying these mutations are fairly homogeneous and typically limited to erythrocytosis with normal to elevated EPO. However, exceptions exist; for example, there is one case with development of concurrent paraganglioma (PHD2-H374R). Analysis of the erythrocytosis-associated PHD2 missense mutations has shown heterogeneous results. Structural studies reveal that mutations can affect different domains of PHD2. Some are close to the hypoxia-inducible transcription factor α/2-oxoglutarate or the iron binding sites for PHD2. In silico studies demonstrate that the mutations do not always affect fully conserved residues. In vitro and in cellulo studies showed varying effects of the mutations, ranging from mild effects to severe loss of function. The exact mechanism of a potential tumor-suppressor role for PHD2 still needs to be elucidated. A knockin mouse model expressing the first reported PHD2-P317R mutation recapitulates the phenotype observed in humans (erythrocytosis with inappropriately normal serum EPO levels) and demonstrates that haploinsufficiency and partial deregulation of PHD2 is sufficient to cause erythrocytosis.
Collapse
Affiliation(s)
- Betty Gardie
- Laboratoire de Génétique Oncologique de l'Ecole Pratique des Hautes Etudes, Villejuif; Unité Mixte de Recherche, Institut national de la santé et de la recherche médicale U892, Centre national de la recherche scientifique 6299, Centre de Recherche en Cancérologie Nantes/Angers, Université de Nantes, Nantes, France
| | - Melanie J Percy
- Department of Haematology, Belfast City Hospital, Belfast, UK
| | - David Hoogewijs
- Institute of Physiology and Zürich Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
| | - Rasheduzzaman Chowdhury
- Department of Chemistry and Oxford Centre for Integrative Systems Biology, Chemistry Research Laboratory, University of Oxford, Oxford, UK
| | - Celeste Bento
- Department of Hematology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Patrick R Arsenault
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stéphane Richard
- Laboratoire de Génétique Oncologique de l'Ecole Pratique des Hautes Etudes, Villejuif; Institut national de la santé et de la recherche médicale U753, Institut de cancérologie Gustave Roussy (IGR), Villejuif, France; Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre, France
| | - Helena Almeida
- Department of Hematology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Frédéric Lambert
- Center for Human Genetics, Pathology Institute, UniLab-Lg, Molecular Haemato-Oncology Unit, CHU of Liege, Liege, Belgium
| | | | - Christopher J Schofield
- Department of Chemistry and Oxford Centre for Integrative Systems Biology, Chemistry Research Laboratory, University of Oxford, Oxford, UK
| | - Frank S Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Rabinowitz MH. Inhibition of hypoxia-inducible factor prolyl hydroxylase domain oxygen sensors: tricking the body into mounting orchestrated survival and repair responses. J Med Chem 2013; 56:9369-402. [PMID: 23977883 DOI: 10.1021/jm400386j] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypoxia-inducible factor (HIF) is an oxygen-sensitive dimeric transcription factor that responds to pathophysiologically low O2 tensions via up-regulation, which leads to an orchestrated biological response to hypoxia. The HIF prolyl hydroxylase domain (PHD) enzymes are non-heme, iron-containing dioxygenases requiring for activity both molecular oxygen and 2-oxoglutarate that, under normoxia, selectively hydroxylate proline residues of HIF, initiating proteosomal degradation of the latter. The dependence of HIF protein levels on the concentration of O2 present, mediated by the PHD enzymes, forms the basis for one of the most significant biological sensor systems of tissue oxygenation in response to ischemic and inflammatory events. Consequently, pharmacological inhibition of PHD enzymes, leading to stabilization of HIF, may be of considerable therapeutic potential in treating conditions of tissue stress and injury. This Perspective reviews the PHDs and small molecule drug discovery efforts. A critical view of this challenging field is offered, which addresses potential concerns and highlights exciting possibilities for the future.
Collapse
Affiliation(s)
- Michael H Rabinowitz
- Janssen Pharmaceutical Research & Development, LLC , 3210 Merryfield Row, San Diego, California 92121, United States
| |
Collapse
|
35
|
Meidan R, Klipper E, Zalman Y, Yalu R. The role of hypoxia-induced genes in ovarian angiogenesis. Reprod Fertil Dev 2013; 25:343-50. [PMID: 22950963 DOI: 10.1071/rd12139] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/26/2012] [Indexed: 01/23/2023] Open
Abstract
The hypoxic microenvironment that occurs in fast-growing tissue such as the corpus luteum (CL) is a major contributor to its ability to survive via the induction of an intricate vascular network. Cellular responses to hypoxia are mediated by hypoxia-inducible factor-1 (HIF-1), an oxygen-regulated transcriptional activator. HIF-1, a heterodimer consisting of a constitutively-expressed β subunit and an oxygen-regulated α subunit, binds to the hypoxia responsive element (HRE) present in the promoter regions of responsive genes. This review summarises evidence for the involvement of hypoxia and HIF-1α in CL development and function. Special emphasis is given to hypoxia-induced, luteal cell-specific expression of multiple genes (vascular endothelial growth factor A (VEGFA), fibroblast growth factor 2 (FGF-2), prokineticin receptor 2 (PK-R2), stanniocalcin 1 (STC-1) and endothelin 2 (EDN-2) that participate in the angiogenic process during CL formation.
Collapse
Affiliation(s)
- Rina Meidan
- Department of Animal Sciences, The Hebrew University of Jerusalem, The Robert H. Smith Faculty of Agriculture, Food and Environment, Rehovot 76100, Israel.
| | | | | | | |
Collapse
|
36
|
Skarzynski DJ, Piotrowska-Tomala KK, Lukasik K, Galvão A, Farberov S, Zalman Y, Meidan R. Growth and Regression in Bovine Corpora Lutea: Regulation by Local Survival and Death Pathways. Reprod Domest Anim 2013; 48 Suppl 1:25-37. [DOI: 10.1111/rda.12203] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 05/20/2013] [Indexed: 11/30/2022]
Affiliation(s)
- DJ Skarzynski
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - KK Piotrowska-Tomala
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - K Lukasik
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - A Galvão
- Department of Reproductive Immunology and Pathology; Institute of Animal Reproduction and Food Research; Polish Academy of Sciences; Olsztyn; Poland
| | - S Farberov
- Department of Animal Sciences; The Robert H. Smith Faculty of Agriculture, Food and Environment; The Hebrew University of Jerusalem; Rehovot; Israel
| | - Y Zalman
- Department of Animal Sciences; The Robert H. Smith Faculty of Agriculture, Food and Environment; The Hebrew University of Jerusalem; Rehovot; Israel
| | - R Meidan
- Department of Animal Sciences; The Robert H. Smith Faculty of Agriculture, Food and Environment; The Hebrew University of Jerusalem; Rehovot; Israel
| |
Collapse
|
37
|
Smirnova NA, Hushpulian DM, Speer RE, Gaisina IN, Ratan RR, Gazaryan IG. Catalytic mechanism and substrate specificity of HIF prolyl hydroxylases. BIOCHEMISTRY (MOSCOW) 2013; 77:1108-19. [PMID: 23157291 DOI: 10.1134/s0006297912100033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review describes the catalytic mechanism, substrate specificity, and structural peculiarities of alpha-ketoglutarate dependent nonheme iron dioxygenases catalyzing prolyl hydroxylation of hypoxia-inducible factor (HIF). Distinct localization and regulation of three isoforms of HIF prolyl hydroxylases suggest their different roles in cells. The recent identification of novel substrates other than HIF, namely β2-adrenergic receptor and the large subunit of RNA polymerase II, places these enzymes in the focus of drug development efforts aimed at development of isoform-specific inhibitors. The challenges and prospects of designing isoform-specific inhibitors are discussed.
Collapse
Affiliation(s)
- N A Smirnova
- Burke Medical Research Institute, White Plains, NY 10605, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Leung IKH, Demetriades M, Hardy AP, Lejeune C, Smart TJ, Szöllössi A, Kawamura A, Schofield CJ, Claridge TDW. Reporter ligand NMR screening method for 2-oxoglutarate oxygenase inhibitors. J Med Chem 2013; 56:547-55. [PMID: 23234607 DOI: 10.1021/jm301583m] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The human 2-oxoglutarate (2OG) dependent oxygenases belong to a family of structurally related enzymes that play important roles in many biological processes. We report that competition-based NMR methods, using 2OG as a reporter ligand, can be used for quantitative and site-specific screening of ligand binding to 2OG oxygenases. The method was demonstrated using hypoxia inducible factor hydroxylases and histone demethylases, and K(D) values were determined for inhibitors that compete with 2OG at the metal center. This technique is also useful as a screening or validation tool for inhibitor discovery, as exemplified by work with protein-directed dynamic combinatorial chemistry.
Collapse
Affiliation(s)
- Ivanhoe K H Leung
- Department of Chemistry, University of Oxford , Chemistry Research Laboratory, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Crestoni ME, Chiavarino B, Scuderi D, Di Marzio A, Fornarini S. Discrimination of 4-hydroxyproline diastereomers by vibrational spectroscopy of the gaseous protonated species. J Phys Chem B 2012; 116:8771-9. [PMID: 22764742 DOI: 10.1021/jp302382p] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydroxylation of proline is a prominent oxidative post-translational modification (oxPTM) in animals, characterized by site specificity and stereochemical control. The presence of this irreversible modification and the ensuing generation of a chiral center have been assayed in (2S,4R)-4-hydroxyproline and (2S,4S)-4-hydroxyproline forming the protonated species by electrospray ionization and sampling them by infrared multiple photon dissociation (IRMPD) spectroscopy. IRMPD spectra, recorded both in the 950-1950 cm(-1) (using the CLIO free electron laser) and in the 3200-3700 cm(-1) [using a tabletop parametric oscillator/amplifier (OPO/OPA) laser] regions, have been interpreted by comparison with the absorbance spectra of the lowest energy structures calculated at MP2/6-311+G** level of theory. Remarkable spectral differences have emerged in the fingerprint region, pointing to the unambiguous discrimination between S,R and S,S diastereomers. The main differences arise from the position of the carbonyl stretching mode, a signature of nonzwitterionic structures, moving from 1750 cm(-1) for the S,R form to 1770 cm(-1) for the S,S diastereomer. Furthermore, a well-defined band associated with the NH(2) wagging mode at 1333 cm(-1) is a distinct mark of the S,S isomer. Each gaseous protonated epimer comprises a population of at least three conformers, stabilized by intramolecular hydrogen bonds linking the two hydrogens of protonated secondary amine group with the 4-hydroxy substituent and with an oxygen atom of the carboxylic group, respectively. Interestingly, a tendency to adopt either C(4)-exo (up) or C(4)-endo (down) pyrrolidine puckering upon proline 4(R)- or 4(S)-hydroxylation, respectively, is observed here. The same bias is found in neutral hydroxyprolines and in collagen model peptides. In the protonated species under examination, this bias originates chirality-induced vibrational features revealed by IRMPD spectroscopy.
Collapse
Affiliation(s)
- Maria Elisa Crestoni
- Dipartimento di Chimica e Tecnologie del Farmaco, Università di Roma La Sapienza, P. le A. Moro 5, I-00185 Roma, Italy.
| | | | | | | | | |
Collapse
|
40
|
Vachal P, Miao S, Pierce JM, Guiadeen D, Colandrea VJ, Wyvratt MJ, Salowe SP, Sonatore LM, Milligan JA, Hajdu R, Gollapudi A, Keohane CA, Lingham RB, Mandala SM, DeMartino JA, Tong X, Wolff M, Steinhuebel D, Kieczykowski GR, Fleitz FJ, Chapman K, Athanasopoulos J, Adam G, Akyuz CD, Jena DK, Lusen JW, Meng J, Stein BD, Xia L, Sherer EC, Hale JJ. 1,3,8-Triazaspiro[4.5]decane-2,4-diones as Efficacious Pan-Inhibitors of Hypoxia-Inducible Factor Prolyl Hydroxylase 1–3 (HIF PHD1–3) for the Treatment of Anemia. J Med Chem 2012; 55:2945-59. [DOI: 10.1021/jm201542d] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Petr Vachal
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Shouwu Miao
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Joan M. Pierce
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Deodial Guiadeen
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Vincent J. Colandrea
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Matthew J. Wyvratt
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Scott P. Salowe
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Lisa M. Sonatore
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - James A. Milligan
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Richard Hajdu
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Anantha Gollapudi
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Carol A. Keohane
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Russell B. Lingham
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Suzanne M. Mandala
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Julie A. DeMartino
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Xinchun Tong
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Michael Wolff
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Dietrich Steinhuebel
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gerard R. Kieczykowski
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Fred J. Fleitz
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Kevin Chapman
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - John Athanasopoulos
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gregory Adam
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Can D. Akyuz
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Dhirendra K. Jena
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Jeffrey W. Lusen
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Juncai Meng
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Benjamin D. Stein
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Lei Xia
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Edward C. Sherer
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Jeffrey J. Hale
- Departments of †Medicinal Chemistry, ‡Infectious Diseases; §Immunology; ∥Drug Metabolism/Pharmacokinetics; ⊥Process Research; #Target Validation; ▽Information Technology; ○Chemistry Modeling
and Informatics. Merck Research Laboratories, Rahway, New Jersey 07065, United States
| |
Collapse
|
41
|
PHD1 interacts with ATF4 and negatively regulates its transcriptional activity without prolyl hydroxylation. Exp Cell Res 2011; 317:2789-99. [PMID: 21951999 DOI: 10.1016/j.yexcr.2011.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Revised: 09/04/2011] [Accepted: 09/09/2011] [Indexed: 12/21/2022]
Abstract
Cellular response to hypoxia plays an important role in both circulatory and pulmonary diseases and cancer. Hypoxia-inducible factors (HIFs) are major transcription factors regulating the response to hypoxia. The α-subunits of HIFs are hydroxylated by members of the prolyl-4-hydroxylase domain (PHD) family, PHD1, PHD2, and PHD3, in an oxygen-dependent manner. Here, we report on the identification of ATF4 as a protein interacting with PHD1 as well as PHD3, but not with PHD2. The central region of ATF4 including the Zipper II domain, ODD domain and β-TrCP recognition motif were involved in the interaction with PHD1. Coexistence of PHD1 stabilized ATF4, as opposed to the destabilization of ATF4 by PHD3. Moreover, coexpression of ATF4 destabilized PHD3, whereas PHD1 stability was not affected by the presence of ATF4. Mutations to alanine of proline residues in ATF4 that satisfied hydroxylation consensus by PHDs did not affect binding activity of ATF4 to PHD1 and PHD3. Furthermore, in vitro prolyl hydroxylation assay clearly indicated that ATF4 did not serve as a substrate of both PHD1 and PHD3. Coexpression of PHD1 or PHD3 with ATF4 repressed the transcriptional activity of ATF4. These results suggest that PHD1 and PHD3 control the transactivation activity of ATF4.
Collapse
|
42
|
van der Wel H, Johnson JM, Xu Y, Karunaratne CV, Wilson KD, Vohra Y, Boons GJ, Taylor CM, Bendiak B, West CM. Requirements for Skp1 processing by cytosolic prolyl 4(trans)-hydroxylase and α-N-acetylglucosaminyltransferase enzymes involved in O₂ signaling in dictyostelium. Biochemistry 2011; 50:1700-13. [PMID: 21247092 PMCID: PMC3192012 DOI: 10.1021/bi101977w] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The social amoeba Dictyostelium expresses a hypoxia inducible factor-α (HIFα) type prolyl 4-hydroxylase (P4H1) and an α-N-acetylglucosaminyltransferase (Gnt1) that sequentially modify proline-143 of Skp1, a subunit of the SCF (Skp1/Cullin/F-box protein) class of E3 ubiquitin ligases. Prior genetic studies have implicated Skp1 and its modification by these enzymes in O(2) regulation of development, suggesting the existence of an ancient O(2)-sensing mechanism related to modification of the transcription factor HIFα by animal prolyl 4-hydroxylases (PHDs). To better understand the role of Skp1 in P4H1-dependent O(2) signaling, biochemical and biophysical studies were conducted to characterize the reaction product and the basis of Skp1 substrate selection by P4H1 and Gnt1. (1)H NMR demonstrated formation of 4(trans)-hydroxyproline as previously found for HIFα, and highly purified P4H1 was inhibited by Krebs cycle intermediates and other compounds that affect animal P4Hs. However, in contrast to hydroxylation of HIFα by PHDs, P4H1 depended on features of full-length Skp1, based on truncation, mutagenesis, and competitive inhibition studies. These features are conserved during animal evolution, as even mammalian Skp1, which lacks the target proline, became a good substrate upon its restoration. P4H1 recognition may depend on features conserved for SCF complex formation as heterodimerization with an F-box protein blocked Skp1 hydroxylation. The hydroxyproline-capping enzyme Gnt1 exhibited similar requirements for Skp1 as a substrate. These and other findings support a model in which the protist P4H1 conditionally hydroxylates Skp1 of E3(SCF)ubiquitin ligases to control half-lives of multiple targets, rather than the mechanism of animal PHDs where individual proteins are hydroxylated leading to ubiquitination by the evolutionarily related E3(VBC)ubiquitin ligases.
Collapse
Affiliation(s)
- Hanke van der Wel
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Jennifer M. Johnson
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Yuechi Xu
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Chamini V. Karunaratne
- Department of Chemistry, 742 Choppin Hall, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Kyle D. Wilson
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Yusuf Vohra
- Dept. of Chemistry and Complex Carbohydrate Research Center, 315 Riverbend Road, University of Georgia, Athens, GA 30602 USA
| | - Geert-Jan Boons
- Dept. of Chemistry and Complex Carbohydrate Research Center, 315 Riverbend Road, University of Georgia, Athens, GA 30602 USA
| | - Carol M. Taylor
- Department of Chemistry, 742 Choppin Hall, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Brad Bendiak
- Department of Cell and Developmental Biology and Structural Biology and Biophysics Program, University of Colorado Denver, Anschutz Medical Campus, Mail Stop 8108, RC-1 South Bldg., L18-12120, 12801 East 17th Avenue, Aurora, CO 80045 USA
| | - Christopher M. West
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| |
Collapse
|
43
|
Hong S, Yum S, Ha NC, Jung Y. von Hippel–Lindau β-domain–luciferase fusion protein as a bioluminescent hydroxyproline sensor for a hypoxia-inducible factor prolyl hydroxylase assay. Anal Biochem 2010; 407:220-5. [DOI: 10.1016/j.ab.2010.08.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 08/05/2010] [Indexed: 01/11/2023]
|
44
|
Smirnova NA, Rakhman I, Moroz N, Basso M, Payappilly J, Kazakov S, Hernandez-Guzman F, Gaisina IN, Kozikowski AP, Ratan RR, Gazaryan IG. Utilization of an in vivo reporter for high throughput identification of branched small molecule regulators of hypoxic adaptation. ACTA ACUST UNITED AC 2010; 17:380-91. [PMID: 20416509 DOI: 10.1016/j.chembiol.2010.03.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 02/26/2010] [Accepted: 03/09/2010] [Indexed: 01/03/2023]
Abstract
Small molecules inhibiting hypoxia inducible factor (HIF) prolyl hydroxylases (PHDs) are the focus of drug development efforts directed toward the treatment of ischemia and metabolic imbalance. A cell-based reporter produced by fusing HIF-1 alpha oxygen degradable domain (ODD) to luciferase was shown to work as a capture assay monitoring stability of the overexpressed luciferase-labeled HIF PHD substrate under conditions more physiological than in vitro test tubes. High throughput screening identified novel catechol and oxyquinoline pharmacophores with a "branching motif" immediately adjacent to a Fe-binding motif that fits selectively into the HIF PHD active site in in silico models. In accord with their structure-activity relationship in the primary screen, the best "hits" stabilize HIF1 alpha, upregulate known HIF target genes in a human neuronal line, and exert neuroprotective effects in established model of oxidative stress in cortical neurons.
Collapse
Affiliation(s)
- Natalya A Smirnova
- Burke Medical Research Institute, Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 785 Mamaroneck Ave, White Plains, NY 10605, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Protein oxidation: role in signalling and detection by mass spectrometry. Amino Acids 2010; 42:5-21. [PMID: 20401673 DOI: 10.1007/s00726-010-0585-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 03/26/2010] [Indexed: 01/10/2023]
Abstract
Proteins can undergo a wide variety of oxidative post-translational modifications (oxPTM); while reversible modifications are thought to be relevant in physiological processes, non-reversible oxPTM may contribute to pathological situations and disease. The oxidant is also important in determining the type of oxPTM, such as oxidation, chlorination or nitration. The best characterized oxPTMs involved in signalling modulation are partial oxidations of cysteine to disulfide, glutathionylated or sulfenic acid forms that can be reversed by thiol reductants. Proline hydroxylation in HIF signalling is also quite well characterized, and there is increasing evidence that specific oxidations of methionine and tyrosine may have some biological roles. For some proteins regulated by cysteine oxidation, the residues and molecular mechanism involved have been extensively studied and are well understood, such as the protein tyrosine phosphatase PTP1B and MAP3 kinase ASK1, as well as transcription factor complex Keap1-Nrf2. The advances in understanding of the role oxPTMs in signalling have been facilitated by advances in analytical technology, in particular tandem mass spectrometry techniques. Combinations of peptide sequencing by collisionally induced dissociation and precursor ion scanning or neutral loss to select for specific oxPTMs have proved very useful for identifying oxidatively modified proteins and mapping the sites of oxidation. The development of specific labelling and enrichment procedures for S-nitrosylation or disulfide formation has proved invaluable, and there is ongoing work to establish analogous methods for detection of nitrotyrosine and other modifications.
Collapse
|
46
|
Kanelakis KC, Palomino HL, Li L, Wu J, Yan W, Rosen MD, Rizzolio MC, Trivedi M, Morton MF, Yang Y, Venkatesan H, Rabinowitz MH, Shankley NP, Barrett TD. Characterization of a Robust Enzymatic Assay for Inhibitors of 2-Oxoglutarate-Dependent Hydroxylases. ACTA ACUST UNITED AC 2009; 14:627-35. [DOI: 10.1177/1087057109333976] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The prolyl-4-hydroxylase proteins regulate the hypoxia-inducible transcription factors (HIFs) by hydroxylation of proline residues targeting HIF-1α for proteasomal degradation. Using the purified catalytic domain of prolyl hydroxylase 2 (PHD2181-417), an enzymatic assay has been developed to test inhibitors of the enzyme in vitro. Because PHD2 hydroxylates HIF-1α, with succinic acid produced as an end product, radiolabeled [5-14C]-2-oxoglutaric acid was used and formation of [14C]-succinic acid was measured to quantify PHD2181-417 enzymatic activity. Comparison of the separation of 2-oxoglutaric acid and succinic acid by either ion exchange chromatography or precipitation with phenylhydrazine showed similar results, but the quantification and throughput were vastly increased using the latter method. The PHD2 reaction was substrate and concentration dependent. The addition of iron to the enzyme reaction mix resulted in an increase in enzymatic activity. The Km value for 2-oxoglutaric acid was determined to be 0.9 µM, and known PHD2 inhibitors were used to validate the assay. In addition, the authors demonstrate that this assay can be applied to other 2-oxoglutaric acid-dependent enzymes, including the asparaginyl hydroxylase, factor-inhibiting HIF-1α (FIH). A concentration-dependent increase in succinic acid production using recombinant FIH enzyme with a synthetic peptide substrate was observed. The authors conclude that a by-product enzyme assay measuring the conversion of 2-oxoglutaric acid to succinic acid using the catalytic domain of the human PHD2 provides a convenient method for the biochemical evaluation of inhibitors of the 2-oxoglutaric acid-dependent hydroxylases. ( Journal of Biomolecular Screening 2009:627-635)
Collapse
Affiliation(s)
- Kimon C. Kanelakis
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California,
| | - Heather L. Palomino
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Lina Li
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Jiejun Wu
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Wen Yan
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Mark D. Rosen
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Michele C. Rizzolio
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Meghana Trivedi
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California, University of California, San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, California
| | - Magda F. Morton
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Young Yang
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Hariharan Venkatesan
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Michael H. Rabinowitz
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Nigel P. Shankley
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| | - Terrance D. Barrett
- Department of Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development L.L.C., San Diego, California
| |
Collapse
|
47
|
The tumor microenvironment and metastatic disease. Clin Exp Metastasis 2008; 26:19-34. [PMID: 18543068 DOI: 10.1007/s10585-008-9182-2] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 05/12/2008] [Indexed: 12/15/2022]
Abstract
The microenvironment of solid tumors is a heterogeneous, complex milieu for tumor growth and survival that includes features such as acidic pH, low nutrient levels, elevated interstitial fluid pressure (IFP) and chronic and fluctuating levels of oxygenation that relate to the abnormal vascular network that exists in tumors. The metastatic potential of tumor cells is believed to be regulated by interactions between the tumor cells and their extracellular environment (extracellular matrix (ECM)). These interactions can be modified by the accumulation of genetic changes and by the transient alterations in gene expression induced by the local tumor microenvironment. Clinical and experimental evidence suggests that altered gene expression in response to the hypoxic microenvironment is a contributing factor to increased metastatic efficiency. A number of genes that have been implicated in the metastatic process, involving angiogenesis, intra/extravasation, survival and growth, have been found to be hypoxia-responsive. The various metastatic determinants, genetic and epigenetic, somatic and inherited may serve as precedents for the future identification of more genes that are involved in metastasis. Much research has focused on genetic and molecular properties of the tumor cells themselves. In the present review we discuss the epigenetic and physiological regulation of metastasis and emphasize the need for further studies on the interactions between the pathophysiologic tumor microenvironment and the tumor extracellular matrix.
Collapse
|
48
|
Alcaide-German ML, Vara-Vega A, Garcia-Fernandez LF, Landazuri MO, del Peso L. A yeast three-hybrid system that reconstitutes mammalian hypoxia inducible factor regulatory machinery. BMC Cell Biol 2008; 9:18. [PMID: 18402654 PMCID: PMC2346465 DOI: 10.1186/1471-2121-9-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Accepted: 04/10/2008] [Indexed: 12/23/2022] Open
Abstract
Background Several human pathologies, including neoplasia and ischemic cardiovascular diseases, course with an unbalance between oxygen supply and demand (hypoxia). Cells within hypoxic regions respond with the induction of a specific genetic program, under the control of the Hypoxia Inducible Factor (HIF), that mediates their adaptation to the lack of oxygen. The activity of HIF is mainly regulated by the EGL-nine homolog (EGLN) enzymes that hydroxylate the alpha subunit of this transcription factor in an oxygen-dependent reaction. Hydroxylated HIF is then recognized and ubiquitinilated by the product of the tumor suppressor gene, pVHL, leading to its proteosomal degradation. Under hypoxia, the hydroxylation of HIF by the EGLNs is compromised due to the lack of oxygen, which is a reaction cosubstrate. Thus, HIF escapes degradation and drives the transcription of its target genes. Since the progression of the aforementioned pathologies might be influenced by activation of HIF-target genes, development of small molecules with the ability to interfere with the HIF-regulatory machinery is of great interest. Results Herein we describe a yeast three-hybrid system that reconstitutes mammalian HIF regulation by the EGLNs and VHL. In this system, yeast growth, under specific nutrient restrictions, is driven by the interaction between the β domain of VHL and a hydroxyproline-containing HIFα peptide. In turn, this interaction is strictly dependent on EGLN activity that hydroxylates the HIFα peptide. Importantly, this system accurately preserves the specificity of the hydroxylation reaction toward specific substrates. We propose that this system, in combination with a matched control, can be used as a simple and inexpensive assay to identify molecules that specifically modulate EGLN activity. As a proof of principle we show that two known EGLN inhibitors, dimethyloxaloylglycine (DMOG) and 6-chlor-3-hydroxychinolin-2-carbonic acid-N-carboxymethylamide (S956711), have a profound and specific effect on the yeast HIF/EGLN/VHL system. Conclusion The system described in this work accurately reconstitutes HIF regulation while preserving EGLN substrate specificity. Thus, it is a valuable tool to study HIF regulation, and particularly EGLN biochemistry, in a cellular context. In addition, we demonstrate that this system can be used to identify specific inhibitors of the EGLN enzymes.
Collapse
Affiliation(s)
- Maria L Alcaide-German
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier 4, 28029 Madrid, Spain.
| | | | | | | | | |
Collapse
|