1
|
Nair P, Sun J, Xie L, Kennedy L, Kozakiewicz D, Kleywegt SM, Hao C, Byun H, Barrett H, Baker J, Monaghan J, Krogh ET, Song D, Peng H. Synthesis and Toxicity Evaluation of p-Phenylenediamine-Quinones. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:7485-7494. [PMID: 40197014 DOI: 10.1021/acs.est.4c12220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
N-(1,3-Dimethylbutyl)-N'-phenyl-p-phenylenediamine-quinone (6PPD-Q), the tire rubber-derived transformation product of 6PPD, was recently discovered to cause the acute mortality of coho salmon (Oncorhynchus kisutch). Aiming to identify potential replacement antiozonants for 6PPD that do not produce toxic quinones, seven PPD-quinones with distinct side chains were synthesized to investigate their structure-related toxicities in vivo using rainbow trout (Oncorhynchus mykiss). While 6PPD-Q exerted high toxicity (96 h LC50 = 0.35 μg/L), toxicity was not observed for six other PPD-quinones despite their similar structures. The fish tissue concentrations of 6PPD-Q after sublethal exposure (0.29 μg/L) were comparable to the other PPD-quinones, which indicated that bioaccumulation levels were not the reason for the selective toxicity of 6PPD-Q. Hydroxylated PPD-quinones were detected as the predominant metabolites in fish tissue. Interestingly, a single major aromatic hydroxylation metabolite was detected for the alternate PPD-quinones, but two abundant OH-6PPD-Q isomers were detected for 6PPD-Q. MS2 spectra confirmed that hydroxylation occurred on the alkyl side chain for one isomer. The structurally selective toxicity of 6PPD-Q was also observed in a coho salmon (CSE-119) cell line, which further supports its intrinsic toxicity. This study reported the selective toxicity of 6PPD-Q and pinpointed the possibility for other PPDs to be applied as potential substitutes of 6PPD.
Collapse
Affiliation(s)
- Pranav Nair
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Jianxian Sun
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Linna Xie
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Lisa Kennedy
- Environmental Sciences and Standards Division, Ontario Ministry of the Environment, Conservation and Parks, Toronto, Ontario M9P 3 V6, Canada
| | - Derek Kozakiewicz
- Environmental Sciences and Standards Division, Ontario Ministry of the Environment, Conservation and Parks, Toronto, Ontario M9P 3 V6, Canada
| | - Sonya M Kleywegt
- Environmental Sciences and Standards Division, Ontario Ministry of the Environment, Conservation and Parks, Toronto, Ontario M9P 3 V6, Canada
| | - Chunyan Hao
- Environmental Sciences and Standards Division, Ontario Ministry of the Environment, Conservation and Parks, Toronto, Ontario M9P 3 V6, Canada
| | - Hannah Byun
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Holly Barrett
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Joshua Baker
- Nautilus Environmental, Burnaby, British Columbia V5A 4N7, Canada
| | - Joseph Monaghan
- Applied Environmental Research Laboratories, Department of Chemistry, Vancouver Island University, Nanaimo, British Columbia V9R 5S5, Canada
| | - Erik T Krogh
- Applied Environmental Research Laboratories, Department of Chemistry, Vancouver Island University, Nanaimo, British Columbia V9R 5S5, Canada
| | - Datong Song
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
- School of the Environment, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
2
|
Wang Y, Wang Y, Hu Y, Wu Q, Gui L, Zeng W, Chen Q, Yu T, Zhang X, Lan K. CYP8B1 Catalyzes 12alpha-Hydroxylation of C 27 Bile Acid: In Vitro Conversion of Dihydroxycoprostanic Acid into Trihydroxycoprostanic Acid. Drug Metab Dispos 2024; 52:1234-1243. [PMID: 39214664 DOI: 10.1124/dmd.124.001694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Sterol 12α-hydroxylase (CYP8B1) is the unique P450 enzyme with sterol 12-oxidation activity, playing an exclusive role in 12α-hydroxylating intermediates along the bile acid (BA) synthesis pathway. Despite the long history of BA metabolism studies, it is unclear whether CYP8B1 catalyzes 12α-hydroxylation of C27 BAs, the key intermediates shuttling between mitochondria and peroxisomes. This work provides robust in vitro evidence that both microsomal and recombinant CYP8B1 enzymes catalyze the 12α-hydroxylation of dihydroxycoprostanic acid (DHCA) into trihydroxycoprostanic acid (THCA). On the one hand, DHCA 12α-hydroxylation reactivity is conservatively detected in liver microsomes of both human and preclinical animals. The reactivity of human tissue fractions conforms well with the selectivity of CYP8B1 mRNA expression, while the contribution of P450 enzymes other than CYP8B1 is excluded by reaction phenotyping in commercial recombinant enzymes. On the other hand, we prepared functional recombinant human CYP8B1 proteins according to a recently published protocol. Titration of the purified CYP8B1 proteins with either C4 (7α-hydroxy-4-cholesten-3-one) or DHCA yields expected blue shifts of the heme Soret peak (type I binding). The recombinant CYP8B1 proteins efficiently catalyze 12α-hydroxylation of both DHCA and C4, with substrate concentration occupying half of the binding sites of 3.0 and 1.9 μM and kcat of 3.2 and 2.6 minutes-1, respectively. In summary, the confirmed role of CYP8B1 in 12α-hydroxylation of C27 BAs has furnished the forgotten passageway in the BA synthesis pathway. The present finding might have opened a new window to consider the biology of CYP8B1 in glucolipid metabolism and to evaluate CYP8B1 inhibition as a therapeutic approach of crucial interest for metabolic diseases. SIGNIFICANCE STATEMENT: The academic community has spent approximately 90 years interpreting the synthesis of bile acids. However, the 12α-hydroxylation of intermediates catalyzed by CYP8B1 is not completely mapped on the classic pathway, particularly for the C27 bile acids, the pivotal intermediates shuttling between mitochondria and peroxisomes. This work discloses the forgotten 12α-hydroxylation pathway from dihydroxycoprostanic acid into trihydroxycoprostanic acid. The present finding may facilitate evaluating CYP8B1 inhibition as a therapeutic approach of crucial interest for metabolic diseases.
Collapse
Affiliation(s)
- Yutong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - Yixuan Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - YiTing Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - QingLiang Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - Lanlan Gui
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - Wushuang Zeng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - Qi Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - Tingting Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - Xinjie Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| | - Ke Lan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China (Yu.W., Yi.W., Y.H., Q.W., L.G., W.Z., Q.C., T.Y., X.Z., K.L.) and Chengdu Cynogen Bio-pharmaceutical Tech. Co., Ltd., Chengdu, China (L.G., W.Z., K.L.)
| |
Collapse
|
3
|
Chaudhary R, Rehman M, Agarwal V, Kumar A, Kaushik AS, Srivastava S, Srivastava S, Verma R, Rajinikanth PS, Mishra V. Terra incognita of glial cell dynamics in the etiology of leukodystrophies: Broadening disease and therapeutic perspectives. Life Sci 2024; 354:122953. [PMID: 39122110 DOI: 10.1016/j.lfs.2024.122953] [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: 02/05/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
4
|
Roumain M, Guillemot-Legris O, Ameraoui H, Alhouayek M, Muccioli GG. Identification and in vivo detection of side-chain hydroxylated metabolites of 4β-hydroxycholesterol. J Steroid Biochem Mol Biol 2023; 234:106376. [PMID: 37604319 DOI: 10.1016/j.jsbmb.2023.106376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
Oxysterols are oxidized derivatives of cholesterol that are formed by enzymatic processes or through the action of reactive oxygen species. Several of these bioactive lipids have been shown to be affected and/or play a role in inflammatory processes. 4β-hydroxycholesterol is one of the major oxysterols in mice and humans and its levels are affected by inflammatory diseases. However, apart from its long half-life, little is known about its catabolism. By incubating 4β-hydroxycholesterol with mouse mitochondria-enriched liver fractions, as well as 25-hydroxycholesterol and 27-hydroxycholesterol with recombinant CYP3A4, we identified 4β,25-dihydroxycholesterol and 4β,27-dihydroxycholesterol as 4β-hydroxycholesterol metabolites. Supporting the biological relevance of this metabolism, we detected both metabolites after incubation of J774, primary mouse peritoneal macrophages and PMA-differentiated THP-1 cells with 4β-hydroxycholesterol. Across our experiments, the incubation of cells with lipopolysaccharides differentially affected the levels of the 25- and 27-hydroxylated metabolites of 4β-hydroxycholesterol. Finally, 4β,27-dihydroxycholesterol was also detected in mice liver and plasma after intraperitoneal administration of 4β-hydroxycholesterol. To our knowledge, this is the first report of the in vitro and in vivo detection and quantification of 4β-hydroxycholesterol metabolites.
Collapse
Affiliation(s)
- Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Hafsa Ameraoui
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Belgium.
| |
Collapse
|
5
|
Wu D, Nealon G, Liu Y, Kim TK, Slominski AT, Tuckey RC. Metabolism of Lumisterol 2 by CYP27A1. J Steroid Biochem Mol Biol 2023; 233:106370. [PMID: 37499840 DOI: 10.1016/j.jsbmb.2023.106370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023]
Abstract
Lumisterol2 (L2) is a photoproduct of UVB action on the fungal membrane sterol, ergosterol. Like vitamin D2, it is present in edible mushrooms, especially after UV irradiation. Lumisterol3 is similarly produced in human skin from 7-dehydrocholesterol by UVB and can be converted to hydroxy-metabolites by CYP27A1 and CYP11A1. These products are biologically active on human cells with actions that include photoprotection and inhibition of proliferation. The aim of this study was to test the ability of CYP11A1 and CYP27A1 to metabolise L2. Purified CYP27A1 was found to efficiently metabolise L2 to three major products and several minor products, whilst CYP11A1 did not act appreciably on L2. The three major products of CYP27A1 action on L2 were identified by mass spectrometry and NMR as 24-hydroxyL2, 27-hydroxyL2 and 28-hydroxyL2. Minor products included two dihydroxy L2 species, one which was identified as 24,27(OH)2L2, and another metabolite with one oxo and one hydroxyl group added. A comparison on the kinetics of the metabolism of L2 by CYP27A1 with that of the structurally similar compounds, L3 and ergosterol, was carried out with substrates incorporated into phospholipid vesicles. CYP27A1 displayed a 12-fold lower Km with L2 as substrate compared to L3 and a 5-fold lower turnover number (kcat), resulting in a 2.2 fold higher catalytic efficiency (kcat/Km) for L2 metabolism. L2 was a much better substrate for CYP27A1 than its precursor, ergosterol, with a catalytic efficiency 18-fold higher. The major CYP27A1-derived hydroxy-L2 products, 24-hydroxyL2, 27-hydroxyL2 and 28-hydroxyL2, inhibited the proliferation of melanoma and epidermoid cancer cell lines. In conclusion, this study shows that L2 is not metabolized appreciably by CYP11A1, but it is a good substrate for CYP27A1 which hydroxylates its side chain to produce 3 major products that display anti-proliferative activity on skin-cancer cell lines.
Collapse
Affiliation(s)
- Dongxian Wu
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Gareth Nealon
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia; Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Perth, WA 6009, Australia
| | - Yuchen Liu
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Tae-Kang Kim
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA; VA Medical Center, Birmingham, AL, USA
| | - Robert C Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia.
| |
Collapse
|
6
|
Norlin M, Wikvall K. Enzymatic activation in vitamin D signaling - Past, present and future. Arch Biochem Biophys 2023; 742:109639. [PMID: 37196753 DOI: 10.1016/j.abb.2023.109639] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/10/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Vitamin D signaling is important in regulating calcium homeostasis essential for bone health but also displays other functions in cells of several tissues. Disturbed vitamin D signaling is linked to a large number of diseases. The multiple cytochrome P450 (CYP) enzymes catalyzing the different hydroxylations in bioactivation of vitamin D3 are crucial for vitamin D signaling and function. This review is focused on the progress achieved in identification of the bioactivating enzymes and their genes in production of 1α,25-dihydroxyvitamin D3 and other active metabolites. Results obtained on species- and tissue-specific expression, catalytic reactions, substrate specificity, enzyme kinetics, and consequences of gene mutations are evaluated. Matters of incomplete understanding regarding the physiological roles of some vitamin D hydroxylases are critically discussed and the authors will give their view of the importance of each enzyme for vitamin D signaling. Roles of different vitamin D receptors and an alternative bioactivation pathway, leading to 20-hydroxylated vitamin D3 metabolites, are also discussed. Considerable progress has been achieved in knowledge of the vitamin D3 bioactivating enzymes. Nevertheless, several intriguing areas deserve further attention to understand the pleiotropic and diverse activities elicited by vitamin D signaling and the mechanisms of enzymatic activation necessary for vitamin D-induced responses.
Collapse
Affiliation(s)
- Maria Norlin
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| | - Kjell Wikvall
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
7
|
Jacob JB, Wei KC, Bepler G, Reyes JD, Cani A, Polin L, White K, Kim S, Viola N, McGrath J, Guastella A, Yin C, Mi QS, Kidder BL, Wagner KU, Ratner S, Phillips V, Xiu J, Parajuli P, Wei WZ. Identification of actionable targets for breast cancer intervention using a diversity outbred mouse model. iScience 2023; 26:106320. [PMID: 36968078 PMCID: PMC10034465 DOI: 10.1016/j.isci.2023.106320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/16/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
HER2-targeted therapy has improved breast cancer survival, but treatment resistance and disease prevention remain major challenges. Genes that enable HER2/Neu oncogenesis are the next intervention targets. A bioinformatics discovery platform of HER2/Neu-expressing Diversity Outbred (DO) F1 Mice was established to identify cancer-enabling genes. Quantitative Trait Loci (QTL) associated with onset ages and growth rates of spontaneous mammary tumors were sought. Twenty-six genes in 3 QTL contain sequence variations unique to the genetic backgrounds that are linked to aggressive tumors and 21 genes are associated with human breast cancer survival. Concurrent identification of TSC22D3, a transcription factor, and its target gene LILRB4, a myeloid cell checkpoint receptor, suggests an immune axis for regulation, or intervention, of disease. We also investigated TIEG1 gene that impedes tumor immunity but suppresses tumor growth. Although not an actionable target, TIEG1 study revealed genetic regulation of tumor progression, forming the basis of the genetics-based discovery platform.
Collapse
Affiliation(s)
- Jennifer B. Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kuang-Chung Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Gerold Bepler
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joyce D. Reyes
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Andi Cani
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lisa Polin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kathryn White
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Seongho Kim
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Nerissa Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Julie McGrath
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Anthony Guastella
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - CongCong Yin
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Qing-Shen Mi
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Benjamin L. Kidder
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kay-Uwe Wagner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Stuart Ratner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Victoria Phillips
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joanne Xiu
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Prahlad Parajuli
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Wei-Zen Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
8
|
Tuckey RC, Cheng CYS, Li L, Jiang Y. Analysis of the ability of vitamin D3-metabolizing cytochromes P450 to act on vitamin D3 sulfate and 25-hydroxyvitamin D3 3-sulfate. J Steroid Biochem Mol Biol 2023; 227:106229. [PMID: 36455719 DOI: 10.1016/j.jsbmb.2022.106229] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/08/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
25-Hydroxyvitamin D3 (25(OH)D3) is present in the human circulation esterified to sulfate with some studies showing that 25(OH)D3 3-sulfate levels are almost as high as unconjugated 25(OH)D3. Vitamin D3 is also present in human serum in the sulfated form as are other metabolites. Our aim was to determine whether sulfated forms of vitamin D3 and vitamin D3 metabolites can be acted on by vitamin D-metabolizing cytochromes P450 (CYPs), one of which (CYP11A1) is known to act on cholesterol sulfate. We used purified, bacterially expressed CYPs to test if they could act on the sulfated forms of their natural substrates. Purified CYP27A1 converted vitamin D3 sulfate to 25(OH)D3 3-sulfate with a catalytic efficiency (kcat/Km) approximately half that for the conversion of vitamin D3 to 25(OH)D3. Similarly, the rate of metabolism of vitamin D3 sulfate was half that of vitamin D3 for CYP27A1 in rat liver mitochondria. CYP2R1 which is also a vitamin D 25-hydroxylase did not act on vitamin D3 sulfate. CYP11A1 was able to convert vitamin D3 sulfate to 20(OH)D3 3-sulfate but at a considerably lower rate than for conversion of vitamin D3 to 20(OH)D3. 25(OH)D3 3-sulfate was not metabolized by the activating enzyme, CYP27B1, nor by the inactivating enzyme, CYP24A1. Thus, we conclude that 25(OH)D3 3-sulfate in the circulation may act as a pool of metabolically inactive vitamin D3 to be released by hydrolysis at times of need whereas vitamin D3 sulfate can be metabolized in a similar manner to free vitamin D3 by CYP27A1 and to a lesser degree by CYP11A1.
Collapse
Affiliation(s)
- Robert C Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia.
| | - Chloe Y S Cheng
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Lei Li
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Yuhan Jiang
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
9
|
Abstract
Most cholesterol in mammalian cells is stored in the plasma membrane (PM). Cholesterol transport from the PM to low-sterol regulatory regions of the endoplasmic reticulum (ER) controls cholesterol synthesis and uptake, and thereby influences the rates of cholesterol flux between tissues of complex organisms. Cholesterol transfer to the ER is also required for steroidogenesis, oxysterol and bile acid synthesis, and cholesterol esterification. The ER-resident Aster proteins (Aster-A, -B, and -C) form contacts with the PM to move cholesterol to the ER in mammals. Mice lacking Aster-B have low adrenal cholesteryl ester stores and impaired steroidogenesis because of a defect in cholesterol transport from high-density lipoprotein (HDL) to the ER. This work reviews the molecular characteristics of Asters, their role in HDL- and low-density lipoprotein (LDL)-cholesterol movement, and how cholesterol transferred to the ER is utilized by cells. The roles of other lipid transporters and of membrane lipid organization in maintaining aspects of cholesterol homeostasis are also highlighted.
Collapse
Affiliation(s)
- John P Kennelly
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Department of Biological Chemistry, Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
10
|
Bile acids and their receptors in regulation of gut health and diseases. Prog Lipid Res 2023; 89:101210. [PMID: 36577494 DOI: 10.1016/j.plipres.2022.101210] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/26/2022]
Abstract
It is well established that bile acids play important roles in lipid metabolism. In recent decades, bile acids have also been shown to function as signaling molecules via interacting with various receptors. Bile acids circulate continuously through the enterohepatic circulation and go through microbial transformation by gut microbes, and thus bile acids metabolism has profound effects on the liver and intestinal tissues as well as the gut microbiota. Farnesoid X receptor and G protein-coupled bile acid receptor 1 are two pivotal bile acid receptors that highly expressed in the intestinal tissues, and they have emerged as pivotal regulators in bile acids metabolism, innate immunity and inflammatory responses. There is considerable interest in manipulating the metabolism of bile acids and the expression of bile acid receptors as this may be a promising strategy to regulate intestinal health and disease. This review aims to summarize the roles of bile acids and their receptors in regulation of gut health and diseases.
Collapse
|
11
|
Frascoli M, Reboldi A, Kang J. Dietary Cholesterol Metabolite Regulation of Tissue Immune Cell Development and Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:645-653. [PMID: 35961669 PMCID: PMC10215006 DOI: 10.4049/jimmunol.2200273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/14/2022] [Indexed: 01/04/2023]
Abstract
Obesity is considered the primary environmental factor associated with morbidity and severity of wide-ranging inflammatory disorders. The molecular mechanism linking high-fat or cholesterol diet to imbalances in immune responses, beyond the increased production of generic inflammatory factors, is just beginning to emerge. Diet cholesterol by-products are now known to regulate function and migration of diverse immune cell subsets in tissues. The hydroxylated metabolites of cholesterol oxysterols as central regulators of immune cell positioning in lymphoid and mucocutaneous tissues is the focus of this review. Dedicated immunocyte cell surface receptors sense spatially distributed oxysterol tissue depots to tune cell metabolism and function, to achieve the "right place at the right time" axiom of efficient tissue immunity.
Collapse
Affiliation(s)
- Michela Frascoli
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|
12
|
Choudhuri S, Klaassen CD. Molecular Regulation of Bile Acid Homeostasis. Drug Metab Dispos 2022; 50:425-455. [PMID: 34686523 DOI: 10.1124/dmd.121.000643] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022] Open
Abstract
Bile acids have been known for decades to aid in the digestion and absorption of dietary fats and fat-soluble vitamins in the intestine. The development of gene knockout mice models and transgenic humanized mouse models have helped us understand other functions of bile acids, such as their role in modulating fat, glucose, and energy metabolism, and in the molecular regulation of the synthesis, transport, and homeostasis of bile acids. The G-protein coupled receptor TGR5 regulates the bile acid induced alterations of intermediary metabolism, whereas the nuclear receptor FXR regulates bile acid synthesis and homeostasis. However, this review indicates that unidentified factors in addition to FXR must exist to aid in the regulation of bile acid synthesis and homeostasis. SIGNIFICANCE STATEMENT: This review captures the present understanding of bile acid synthesis, the role of bile acid transporters in the enterohepatic circulation of bile acids, the role of the nuclear receptor FXR on the regulation of bile acid synthesis and bile acid transporters, and the importance of bile acids in activating GPCR signaling via TGR5 to modify intermediary metabolism. This information is useful for developing drugs for the treatment of various hepatic and intestinal diseases, as well as the metabolic syndrome.
Collapse
Affiliation(s)
- Supratim Choudhuri
- Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland (S.C.) and Department of Pharmacology, Toxicology, and Therapeutics, School of Medicine, University of Kansas, Kansas City, Kansas (C.D.K.)
| | - Curtis D Klaassen
- Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland (S.C.) and Department of Pharmacology, Toxicology, and Therapeutics, School of Medicine, University of Kansas, Kansas City, Kansas (C.D.K.)
| |
Collapse
|
13
|
Green MR, Sambrook J. Preparation of Labeled DNA, RNA, and Oligonucleotide Probes. Cold Spring Harb Protoc 2022; 2022:2022/1/pdb.top100578. [PMID: 34983861 DOI: 10.1101/pdb.top100578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Labeled nucleic acids and oligonucleotides are typically generated by enzymatic methods such as end-labeling, random priming, nick translation, in vitro transcription, and variations of the polymerase chain reaction (PCR). Some of these methods place the label in specific locations within the nucleic acid (e.g., at the 5' or 3' terminus); others generate molecules that are labeled internally at multiple sites. Some methods yield labeled single-stranded products, whereas others generate double-stranded nucleic acids. Finally, some generate probes of defined length, whereas others yield a heterogeneous population of labeled molecules. Options available for generating and detecting labeled nucleic acids, as well as advice on designing oligonucleotides for use as probes, is included here.
Collapse
|
14
|
Thorne JL, Cioccoloni G. Nuclear Receptors and Lipid Sensing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:83-105. [DOI: 10.1007/978-3-031-11836-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
15
|
Poniedziałek-Czajkowska E, Mierzyński R. Could Vitamin D Be Effective in Prevention of Preeclampsia? Nutrients 2021; 13:nu13113854. [PMID: 34836111 PMCID: PMC8621759 DOI: 10.3390/nu13113854] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022] Open
Abstract
Prevention of preeclampsia (PE) remains one of the most significant problems in perinatal medicine. Due to the possible unpredictable course of hypertension in pregnancy, primarily PE and the high complication rate for the mother and fetus/newborn, it is urgent to offer pregnant women in high-risk groups effective methods of preventing the PE development or delaying its appearance. In addition, due to the association of PE with an increased risk of developing cardiovascular diseases (CVD) in later life, effective preeclampsia prevention could also be important in reducing their incidence. Ideal PE prophylaxis should target the pathogenetic changes leading to the development of PE and be safe for the mother and fetus, inexpensive and freely available. Currently, the only recognized method of PE prevention recommended by many institutions around the world is the use of a small dose of acetylsalicylic acid in pregnant women with risk factors. Unfortunately, some cases of PE are diagnosed in women without recognized risk factors and in those in whom prophylaxis with acetylsalicylic acid is not adequate. Hence, new drugs which would target pathogenetic elements in the development of preeclampsia are studied. Vitamin D (Vit D) seems to be a promising agent due to its beneficial effect on placental implantation, the immune system, and angiogenic factors. Studies published so far emphasize the relationship of its deficiency with the development of PE, but the data on the benefits of its supplementation to reduce the risk of PE are inconclusive. In the light of current research, the key issue is determining the protective concentration of Vit D in a pregnant woman. The study aims to present the possibility of using Vit D to prevent PE, emphasizing its impact on the pathogenetic elements of preeclampsia development.
Collapse
|
16
|
Misra S, Ghatak S, Moreno-Rodriguez RA, Norris RA, Hascall VC, Markwald RR. Periostin/Filamin-A: A Candidate Central Regulatory Axis for Valve Fibrogenesis and Matrix Compaction. Front Cell Dev Biol 2021; 9:649862. [PMID: 34150753 PMCID: PMC8209548 DOI: 10.3389/fcell.2021.649862] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Discoveries in the identification of transcription factors, growth factors and extracellular signaling molecules have led to the detection of downstream targets that modulate valvular tissue organization that occurs during development, aging, or disease. Among these, matricellular protein, periostin, and cytoskeletal protein filamin A are highly expressed in developing heart valves. The phenotype of periostin null indicates that periostin promotes migration, survival, and differentiation of valve interstitial cushion cells into fibroblastic lineages necessary for postnatal valve remodeling/maturation. Genetically inhibiting filamin A expression in valve interstitial cushion cells mirrored the phenotype of periostin nulls, suggesting a molecular interaction between these two proteins resulted in poorly remodeled valve leaflets that might be prone to myxomatous over time. We examined whether filamin A has a cross-talk with periostin/signaling that promotes remodeling of postnatal heart valves into mature leaflets. RESULTS We have previously shown that periostin/integrin-β1 regulates Pak1 activation; here, we revealed that the strong interaction between Pak1 and filamin A proteins was only observed after stimulation of VICs with periostin; suggesting that periostin/integrin-β-mediated interaction between FLNA and Pak1 may have a functional role in vivo. We found that FLNA phosphorylation (S2152) is activated by Pak1, and this interaction was observed after stimulation with periostin/integrin-β1/Cdc42/Rac1 signaling; consequently, FLNA binding to Pak1 stimulates its kinase activity. Patients with floppy and/or prolapsed mitral valves, when genetically screened, were found to have point mutations in the filamin A gene at P637Q and G288R. Expression of either of these filamin A mutants failed to increase the magnitude of filamin A (S2152) expression, Pak1-kinase activity, actin polymerization, and differentiation of VICs into mature mitral valve leaflets in response to periostin signaling. CONCLUSION PN-stimulated bidirectional interaction between activated FLNA and Pak1 is essential for actin cytoskeletal reorganization and the differentiation of immature VICs into mature valve leaflets.
Collapse
Affiliation(s)
- Suniti Misra
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Shibnath Ghatak
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Ricardo A. Moreno-Rodriguez
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Vincent C. Hascall
- Department of Biomedical Engineering/ND20, Cleveland Clinic, Cleveland, OH, United States
| | - Roger R. Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
17
|
Rashvand Z, Kahrizi K, Najmabadi H, Najafipour R, Omrani MD. Clinical and Genetic Characteristics of Splicing Variant in CYP27A1 in an Iranian Family with Cerebrotendinous Xanthomatosis. IRANIAN BIOMEDICAL JOURNAL 2021; 25:132-9. [PMID: 33400472 PMCID: PMC7921520 DOI: 10.29252/ibj.25.2.132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background: CTX is a rare congenital lipid-storage disorder, leading to a progressive multisystem disease. CTX with autosomal recessive inheritance is caused by a defect in the CYP27A1 gene. Chronic diarrhea, tendon xanthomas, neurologic impairment, and bilateral cataracts are common symptoms of the disease. Methods: Three affected siblings with an initial diagnosis of non-syndromic intellectual disability were recruited for further molecular investigations. To identify the possible genetic cause(s), WES was performed on the proband. Sanger sequencing was applied to confirm the final variant. The clinical and molecular genetic features of the three siblings from the new CTX family and other patients with the same mutations, as previously reported, were analyzed. The CYP27A1 gene was also studied for the number of pathogenic variants and their location. Results: We found a homozygous splicing mutation, NM_000784: exon6: c.1184+1G>A, in CYP27A1 gene, which was confirmed by Sanger sequencing. Among the detected pathogenic variants, the splice site mutation had the highest prevalence, and the mutations were mostly found in exon 4. Conclusion: This study is the first to report the c.1184+1G>A mutation in Iran. Our findings highlight the other feature of the disease, which is the lack of relationship between phenotype and genotype. Due to nonspecific symptoms and delay in diagnosis, CYP27A1 genetic analysis should be the definitive method for CTX diagnosis.
Collapse
Affiliation(s)
- Zahra Rashvand
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Kahrizi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Reza Najafipour
- Cellular and Molecular Research Centre, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Negri M, Gentile A, de Angelis C, Montò T, Patalano R, Colao A, Pivonello R, Pivonello C. Vitamin D-Induced Molecular Mechanisms to Potentiate Cancer Therapy and to Reverse Drug-Resistance in Cancer Cells. Nutrients 2020; 12:nu12061798. [PMID: 32560347 PMCID: PMC7353389 DOI: 10.3390/nu12061798] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Increasing interest in studying the role of vitamin D in cancer has been provided by the scientific literature during the last years, although mixed results have been reported. Vitamin D deficiency has been largely associated with various types of solid and non-solid human cancers, and the almost ubiquitous expression of vitamin D receptor (VDR) has always led to suppose a crucial role of vitamin D in cancer. However, the association between vitamin D levels and the risk of solid cancers, such as colorectal, prostate and breast cancer, shows several conflicting results that raise questions about the use of vitamin D supplements in cancer patients. Moreover, studies on vitamin D supplementation do not always show improvements in tumor progression and mortality risk, particularly for prostate and breast cancer. Conversely, several molecular studies are in agreement about the role of vitamin D in inhibiting tumor cell proliferation, growth and invasiveness, cell cycle arrest and inflammatory signaling, through which vitamin D may also regulate cancer microenvironment through the activation of different molecular pathways. More recently, a role in the regulation of cancer stem cells proliferation and short non-coding microRNA (miRNAs) expression has emerged, conferring to vitamin D a more crucial role in cancer development and progression. Interestingly, it has been shown that vitamin D is able not only to potentiate the effects of traditional cancer therapy but can even contribute to overcome the molecular mechanisms of drug resistance—often triggering tumor-spreading. At this regard, vitamin D can act at various levels through the regulation of growth of cancer stem cells and the epithelial–mesenchymal transition (EMT), as well as through the modulation of miRNA gene expression. The current review reconsiders epidemiological and molecular literature concerning the role of vitamin D in cancer risk and tumor development and progression, as well as the action of vitamin D supplementation in potentiating the effects of drug therapy and overcoming the mechanisms of resistance often triggered during cancer therapies, by critically addressing strengths and weaknesses of available data from 2010 to 2020.
Collapse
Affiliation(s)
- Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
| | - Annalisa Gentile
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
| | - Cristina de Angelis
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
| | - Tatiana Montò
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
| | - Roberta Patalano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
- Dipartimento di Sanità Pubblica, Università Federico II di Napoli, 80131 Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
- Unesco Chair for Health Education and Sustainable Development, Federico II University, 80131 Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
| | - Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy; (M.N.); (A.G.); (C.d.A.); (T.M.); (R.P.); (A.C.); (R.P.)
- Correspondence:
| |
Collapse
|
19
|
Chockalingam K, Peng Z, Vuong CN, Berghman LR, Chen Z. Golden Gate assembly with a bi-directional promoter (GBid): A simple, scalable method for phage display Fab library creation. Sci Rep 2020; 10:2888. [PMID: 32076016 PMCID: PMC7031318 DOI: 10.1038/s41598-020-59745-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/03/2020] [Indexed: 11/09/2022] Open
Abstract
Fabs offer an attractive platform for monoclonal antibody discovery/engineering, but library construction can be cumbersome. We report a simple method – Golden Gate assembly with a bi-directional promoter (GBid) – for constructing phage display Fab libraries. In GBid, the constant domains of the Fabs are located in the backbone of the phagemid vector and the library insert comprises only the variable regions of the antibodies and a central bi-directional promoter. This vector design reduces the process of Fab library construction to “scFv-like” simplicity and the double promoter ensures robust expression of both constituent chains. To maximize the library size, the 3 fragments comprising the insert – two variable chains and one bi-directional promoter – are assembled via a 3-fragment overlap extension PCR and the insert is incorporated into the vector via a high-efficiency one-fragment, one-pot Golden Gate assembly. The reaction setup requires minimal preparatory work and enzyme quantities, making GBid highly scalable. Using GBid, we constructed a chimeric chicken-human Fab phage display library comprising 1010 variants targeting the multi-transmembrane protein human CD20 (hCD20). Selection/counter-selection on transfected whole cells yielded hCD20-specific antibodies in four rounds of panning. The simplicity and scalability of GBid makes it a powerful tool for the discovery/engineering of Fabs and IgGs.
Collapse
Affiliation(s)
- Karuppiah Chockalingam
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, 77843, USA
| | - Zeyu Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, 77843, USA.,Biosion, Inc., Nanjing, 210061, China
| | - Christine N Vuong
- Department of Poultry Science, Texas A&M University, College Station, Texas, 77843, USA.,Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, 72703, USA
| | - Luc R Berghman
- Department of Poultry Science, Texas A&M University, College Station, Texas, 77843, USA
| | - Zhilei Chen
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, 77843, USA.
| |
Collapse
|
20
|
Majhi PD, Sharma A, Roberts AL, Daniele E, Majewski AR, Chuong LM, Black AL, Vandenberg LN, Schneider SS, Dunphy KA, Jerry DJ. Effects of Benzophenone-3 and Propylparaben on Estrogen Receptor-Dependent R-Loops and DNA Damage in Breast Epithelial Cells and Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:17002. [PMID: 31939680 PMCID: PMC7015622 DOI: 10.1289/ehp5221] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
BACKGROUND Endocrine-disrupting chemicals have been shown to have broad effects on development, but their mutagenic actions that can lead to cancer have been less clearly demonstrated. Physiological levels of estrogen have been shown to stimulate DNA damage in breast epithelial cells through mechanisms mediated by estrogen-receptor alpha (ERα). Benzophenone-3 (BP-3) and propylparaben (PP) are xenoestrogens found in the urine of >96% of U.S. OBJECTIVES We investigated the effect of BP-3 and PP on estrogen receptor-dependent transactivation and DNA damage at concentrations relevant to exposures in humans. METHODS In human breast epithelial cells, DNA damage following treatment with 17β-estradiol (E2), BP-3, and PP was determined by immunostaining with antibodies against γ-H2AX and 53BP1. Estrogenic responses were determined using luciferase reporter assays and gene expression. Formation of R-loops was determined with DNA: RNA hybrid-specific S9.6 antibody. Short-term exposure to the chemicals was also studied in ovariectomized mice. Immunostaining of mouse mammary epithelium was performed to quantify R-loops and DNA damage in vivo. RESULTS Concentrations of 1μM and 5μM BP-3 or PP increased DNA damage similar to that of E2 treatment in a ERα-dependent manner. However, BP-3 and PP had limited transactivation of target genes at 1μM and 5μM concentrations. BP-3 and PP exposure caused R-loop formation in a normal human breast epithelial cell line when ERα was introduced. R-loops and DNA damage were also detected in mammary epithelial cells of mice treated with BP-3 and PP. CONCLUSIONS Acute exposure to xenoestrogens (PP and BP-3) in mice induce DNA damage mediated by formation of ERα-dependent R-loops at concentrations 10-fold lower than those required for transactivation. Exposure to these xenoestrogens may cause deleterious estrogenic responses, such as DNA damage, in susceptible individuals. https://doi.org/10.1289/EHP5221.
Collapse
Affiliation(s)
- Prabin Dhangada Majhi
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Botany, Ravenshaw University, Cuttack, Odisha, India
| | - Aman Sharma
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Amy L. Roberts
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Elizabeth Daniele
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Aliza R. Majewski
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Lynn M. Chuong
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Amye L. Black
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Laura N. Vandenberg
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Sallie S. Schneider
- University of Massachusetts Medical School, Baystate Campus, Springfield, Massachusetts, USA
- Pioneer Valley Life Sciences Institute, Springfield, Massachusetts, USA
| | - Karen A. Dunphy
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - D. Joseph Jerry
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
- Pioneer Valley Life Sciences Institute, Springfield, Massachusetts, USA
| |
Collapse
|
21
|
Ticho AL, Malhotra P, Dudeja PK, Gill RK, Alrefai WA. Intestinal Absorption of Bile Acids in Health and Disease. Compr Physiol 2019; 10:21-56. [PMID: 31853951 PMCID: PMC7171925 DOI: 10.1002/cphy.c190007] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intestinal reclamation of bile acids is crucial for the maintenance of their enterohepatic circulation. The majority of bile acids are actively absorbed via specific transport proteins that are highly expressed in the distal ileum. The uptake of bile acids by intestinal epithelial cells modulates the activation of cytosolic and membrane receptors such as the farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (GPBAR1), which has a profound effect on hepatic synthesis of bile acids as well as glucose and lipid metabolism. Extensive research has focused on delineating the processes of bile acid absorption and determining the contribution of dysregulated ileal signaling in the development of intestinal and hepatic disorders. For example, a decrease in the levels of the bile acid-induced ileal hormone FGF15/19 is implicated in bile acid-induced diarrhea (BAD). Conversely, the increase in bile acid absorption with subsequent overload of bile acids could be involved in the pathophysiology of liver and metabolic disorders such as fatty liver diseases and type 2 diabetes mellitus. This review article will attempt to provide a comprehensive overview of the mechanisms involved in the intestinal handling of bile acids, the pathological implications of disrupted intestinal bile acid homeostasis, and the potential therapeutic targets for the treatment of bile acid-related disorders. Published 2020. Compr Physiol 10:21-56, 2020.
Collapse
Affiliation(s)
- Alexander L. Ticho
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Pooja Malhotra
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Pradeep K. Dudeja
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Ravinder K. Gill
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Waddah A. Alrefai
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- jesse Brown VA Medical Center, Chicago, Illinois, USA
| |
Collapse
|
22
|
Phelps T, Snyder E, Rodriguez E, Child H, Harvey P. The influence of biological sex and sex hormones on bile acid synthesis and cholesterol homeostasis. Biol Sex Differ 2019; 10:52. [PMID: 31775872 PMCID: PMC6880483 DOI: 10.1186/s13293-019-0265-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity and elevated serum lipids are associated with a threefold increase in the risk of developing atherosclerosis, a condition that underlies stroke, myocardial infarction, and sudden cardiac death. Strategies that aim to reduce serum cholesterol through modulation of liver enzymes have been successful in decreasing the risk of developing atherosclerosis and reducing mortality. Statins, which inhibit cholesterol biosynthesis in the liver, are considered among the most successful compounds developed for the treatment of cardiovascular disease. However, recent debate surrounding their effectiveness and safety prompts consideration of alternative cholesterol-lowering therapies, including increasing cholesterol catabolism through bile acid (BA) synthesis. Targeting the enzymes that convert cholesterol to BAs represents a promising alternative to other cholesterol-lowering approaches that treat atherosclerosis as well as fatty liver diseases and diabetes mellitus. Compounds that modify the activity of these pathways have been developed; however, there remains a lack of consideration of biological sex. This is necessary in light of strong evidence for sexual dimorphisms not only in the incidence and progression of the diseases they influence but also in the expression and activity of the proteins affected and in the manner in which men and women respond to drugs that modify lipid handling in the liver. A thorough understanding of the enzymes involved in cholesterol catabolism and modulation by biological sex is necessary to maximize their therapeutic potential.
Collapse
Affiliation(s)
- Taylor Phelps
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Erin Snyder
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Erin Rodriguez
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Hailey Child
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Pamela Harvey
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO, 80309, USA.
| |
Collapse
|
23
|
Petrov AM, Pikuleva IA. Cholesterol 24-Hydroxylation by CYP46A1: Benefits of Modulation for Brain Diseases. Neurotherapeutics 2019; 16:635-648. [PMID: 31001737 PMCID: PMC6694357 DOI: 10.1007/s13311-019-00731-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cholesterol 24-hydroxylation is the major mechanism for cholesterol removal from the brain and the reaction catalyzed by cytochrome P450 46A1 (CYP46A1), a CNS-specific enzyme. This review describes CYP46A1 in the context of cholesterol homeostasis in the brain and summarizes available experimental data on CYP46A1 association with different neurologic diseases, including the mechanisms by which changes in the CYP46A1 activity in the brain could be beneficial for these diseases. The modulation of CYP46A1 activity by genetic and pharmacologic means is also presented along with a brief synopsis of the two clinical trials that evaluate CYP46A1 as a therapeutic target for Alzheimer's disease as well as Dravet and Lennox-Gastaut syndromes.
Collapse
Affiliation(s)
- Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA.
| |
Collapse
|
24
|
Shahoei SH, Nelson ER. Nuclear receptors, cholesterol homeostasis and the immune system. J Steroid Biochem Mol Biol 2019; 191:105364. [PMID: 31002862 PMCID: PMC6589364 DOI: 10.1016/j.jsbmb.2019.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/30/2022]
Abstract
Cholesterol is essential for maintaining membrane fluidity in eukaryotes. Additionally, the synthetic cascade of cholesterol results in precursor molecules important for cellular function such as lipid raft formation and protein prenylation. As such, cholesterol homeostasis is tightly regulated. Interestingly, it is now known that some cholesterol precursors and many metabolites serve as active signaling molecules, binding to different classes of receptors including the nuclear receptors. Furthermore, many cholesterol metabolites or their nuclear receptors have been implicated in the regulation of the immune system in normal physiology and disease. Therefore, in this focused review, cholesterol homeostasis and nuclear receptors involved in this regulation will be discussed, with particular emphasis on how these cascades influence the immune system.
Collapse
Affiliation(s)
- Sayyed Hamed Shahoei
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, United States; Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, IL, United States.
| |
Collapse
|
25
|
Loera-Valencia R, Goikolea J, Parrado-Fernandez C, Merino-Serrais P, Maioli S. Alterations in cholesterol metabolism as a risk factor for developing Alzheimer's disease: Potential novel targets for treatment. J Steroid Biochem Mol Biol 2019; 190:104-114. [PMID: 30878503 DOI: 10.1016/j.jsbmb.2019.03.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and it is characterized by the deposition of amyloid-β (Aβ) plaques and neurofibrillary tangles in the brain. However, the complete pathogenesis of the disease is still unknown. High level of serum cholesterol has been found to positively correlate with an increased risk of dementia and some studies have reported a decreased prevalence of AD in patients taking cholesterol-lowering drugs. Years of research have shown a strong correlation between blood hypercholesterolemia and AD, however cholesterol is not able to cross the Blood Brain Barrier (BBB) into the brain. Cholesterol lowering therapies have shown mixed results in cognitive performance in AD patients, raising questions of whether brain cholesterol metabolism in the brain should be studied separately from peripheral cholesterol metabolism and what their relationship is. Unlike cholesterol, oxidized cholesterol metabolites known as oxysterols are able to cross the BBB from the circulation into the brain and vice-versa. The main oxysterols present in the circulation are 24S-hydroxycholesterol and 27-hydroxycholesterol. These oxysterols and their catalysing enzymes have been found to be altered in AD brains and there is evidence indicating their influence in the progression of the disease. This review gives a broad perspective on the relationship between hypercholesterolemia and AD, cholesterol lowering therapies for AD patients and the role of oxysterols in pathological and non-pathological conditions. Also, we propose cholesterol metabolites as valuable targets for prevention and alternative AD treatments.
Collapse
Affiliation(s)
- Raúl Loera-Valencia
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden.
| | - Julen Goikolea
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - Cristina Parrado-Fernandez
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden; Institute of Molecular Biology and Genetics-IBGM, (University of Valladolid-CSIC), Valladolid, Spain
| | - Paula Merino-Serrais
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden; Instituto Cajal (CSIC), Laboratorio Cajal de Circuitos Corticales, Madrid, Spain
| | - Silvia Maioli
- Karolinska Institutet, Center for Alzheimer Research, Department of Neurobiology Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden.
| |
Collapse
|
26
|
Vps11 and Vps18 of Vps-C membrane traffic complexes are E3 ubiquitin ligases and fine-tune signalling. Nat Commun 2019; 10:1833. [PMID: 31015428 PMCID: PMC6478910 DOI: 10.1038/s41467-019-09800-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 04/02/2019] [Indexed: 12/11/2022] Open
Abstract
In response to extracellular signals, many signalling proteins associated with the plasma membrane are sorted into endosomes. This involves endosomal fusion, which depends on the complexes HOPS and CORVET. Whether and how their subunits themselves modulate signal transduction is unknown. We show that Vps11 and Vps18 (Vps11/18), two common subunits of the HOPS/CORVET complexes, are E3 ubiquitin ligases. Upon overexpression of Vps11/Vps18, we find perturbations of ubiquitination in signal transduction pathways. We specifically demonstrate that Vps11/18 regulate several signalling factors and pathways, including Wnt, estrogen receptor α (ERα), and NFκB. For ERα, we demonstrate that the Vps11/18-mediated ubiquitination of the scaffold protein PELP1 impairs the activation of ERα by c-Src. Thus, proteins involved in membrane traffic, in addition to performing their well-described role in endosomal fusion, fine-tune signalling in several different ways, including through ubiquitination.
Collapse
|
27
|
Abstract
On January 21, 2017, I received an E-mail from Herb Tabor that I had been simultaneously hoping for and dreading for several years: an invitation to write a "Reflections" article for the Journal of Biological Chemistry On the one hand, I was honored to receive an invitation from Herb, a man I have admired for over 40 years, known for 24 years, and worked with as a member of the Editorial Board and Associate Editor of the Journal of Biological Chemistry for 17 years. On the other hand, the invitation marked the waning of my career as an academic scientist. With these conflicting emotions, I wrote this article with the goals of recording my career history and recognizing the many mentors, trainees, and colleagues who have contributed to it and, perhaps with pretension, with the desire that students who are beginning a career in research will find inspiration in the path I have taken and appreciate the importance of luck.
Collapse
Affiliation(s)
- David W Russell
- From the Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9046
| |
Collapse
|
28
|
Ghatak S, Misra S, Moreno-Rodrigue RA, Hascall VC, Leone GW, Markwald RR. Periostin/β1integrin interaction regulates p21-activated kinases in valvular interstitial cell survival and in actin cytoskeleton reorganization. Biochim Biophys Acta Gen Subj 2019; 1863:813-829. [PMID: 30742951 DOI: 10.1016/j.bbagen.2018.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/24/2022]
Abstract
The matricellular protein periostin (PN) promotes postnatal valve remodeling and maturation. Incomplete remodeling of the valve can trigger degenerative processes that lead to a myxomatous phenotype that includes loss of PN. However, signaling pathways involved that link valvular-interstitial-fibroblast cells (VICs) to proliferation, migration and actin remodeling functions are unclear. The p21-activated kinases (Paks) have been shown to regulate cytoskeleton rearrangements and cell proliferation/adhesion/migration functions in a variety of cellular contexts, including normal cells and cancer cells. This study shows that Pak1, but not Pak2 and Pak4, is a critical mediator of VIC survival and actin organization, and that the molecular signaling regulating actin-remodeling is initiated upon PN/beta-integrin-induced phosphorylation of the focal-adhesion-kinase (Fak) (Y397). Molecular and pharmacological inhibition of key components of PN/Fak/Akt1 signaling abolished the PN-induced actin polymerization and the activation of mTOR, p70S6K and Pak1. Similarly, blocking mTOR inhibited p70S6K, Pak1 phosphorylation and consequently actin-polymerization. Accordingly, inhibiting p70S6K blocked Pak1 phosphorylation and actin polymerization, and subsequently inhibited adhesion and growth of VICs. Periostin-induced Akt1 activation of Pak1 is independent of Cdc42 and Rac1 GTPases, and Akt1 is both downstream and upstream of Pak1. Further, the PN-Pak1-induced Akt1 protects cells from apoptosis through suppression of transcriptional activation of Forkhead-Transcription-Factor (FKHR). In contrast, kinase deficient Pak1 increases apoptosis by increasing FKHR-mediated transcriptional activation. These studies define new functional significance of PN-Fak-Akt1-Pak1 signaling that at least partly regulates Akt1-induced actin polymerization and FKHR-mediated transcriptional activation, which may eventually regulate the mature-valve-leaflet remodeling function, and also FKHR-mediated transcriptional activation for pro-survival of VICs.
Collapse
Affiliation(s)
- Shibnath Ghatak
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.; Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St, Charleston, SC 29425, USA
| | - Suniti Misra
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.; Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St, Charleston, SC 29425, USA.
| | - Ricardo A Moreno-Rodrigue
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering/ND20, Cleveland Clinic, Cleveland, OH, USA
| | - Gustavo W Leone
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St, Charleston, SC 29425, USA
| | - Roger R Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA..
| |
Collapse
|
29
|
Holy P, Kloudova A, Soucek P. Importance of genetic background of oxysterol signaling in cancer. Biochimie 2018; 153:109-138. [DOI: 10.1016/j.biochi.2018.04.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/27/2018] [Indexed: 12/14/2022]
|
30
|
Abstract
PURPOSE OF REVIEW Herein, we review the role of FXR and TGR5 in the regulation of hepatic bile acid metabolism, with a focus on how our understanding of bile acid metabolic regulation by these receptors has evolved in recent years and how this improved understanding may facilitate targeting bile acids for type 2 diabetes treatment. RECENT FINDINGS Bile acid profile is a key regulator of metabolic homeostasis. Inhibition of expression of the enzyme that is required for cholic acid synthesis and thus determines bile acid profile, Cyp8b1, may be an effective target for type 2 diabetes treatment. FXR and, more recently, TGR5 have been shown to regulate bile acid metabolism and Cyp8b1 expression and, therefore, may provide a mechanism with which to target bile acid profile for type 2 diabetes treatment. Inhibition of Cyp8b1 expression is a promising therapeutic modality for type 2 diabetes; however, further work is needed to fully understand the pathways regulating Cyp8b1 expression.
Collapse
Affiliation(s)
- Karolina E Zaborska
- Department of Biomedical Sciences, Cornell University, T3 014A Veterinary Research Tower, Ithaca, NY, 14853, USA
| | - Bethany P Cummings
- Department of Biomedical Sciences, Cornell University, T3 014A Veterinary Research Tower, Ithaca, NY, 14853, USA.
| |
Collapse
|
31
|
Karras SN, Wagner CL, Castracane VD. Understanding vitamin D metabolism in pregnancy: From physiology to pathophysiology and clinical outcomes. Metabolism 2018; 86:112-123. [PMID: 29066285 DOI: 10.1016/j.metabol.2017.10.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
This critical time frame of intrauterine life development is considered of major importance on the metabolic imprinting of overall health of the offspring, in later life. This requires a delicate immune balance that nurtures the allogeneic fetus, while maintaining reactivity against pathogens. Dysregulation of these tightly controlled biophenomena at a systemic and placental level, have been considered as a potential mechanism mediating pathogenesis of preeclampsia and spontaneous birth. In this context, vitamin D has been considered as a significant regulator of both innate and adaptive immunity by regulating cell proliferation, differentiation and apoptosis. Vitamin D metabolism during pregnancy manifests striking differences as compared to the non-pregnant state. Calcitriol is increasing >2-3 fold in the first weeks of pregnancy whereas maternal 25-hydroxyvitamin D crosses the placental barrier and represents the main pool of vitamin D in the fetus. Moreover, during pregnancy, vitamin D receptor and regulatory metabolic enzymes are expressed in the placenta and decidua, indicating a potential critical point in the immunomodulation at the maternal-fetal interface. Considering these effects, maternal hypovitaminosis D during pregnancy has been associated with pregnancy related disorders. This review focuses on the mechanistic basis of these adaptive changes, as a background for the development of pregnancy related disorders, with a discourse on the pathophysiology relating hypovitaminosis D and clinical outcomes.
Collapse
Affiliation(s)
- Spyridon N Karras
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece.
| | - Carol L Wagner
- Division of Neonatology, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - V Daniel Castracane
- Department of Obstetrics and Gynecology, Texas Tech University School of Medicine, Odessa, TX, United States
| |
Collapse
|
32
|
Tuckey RC, Li W, Ma D, Cheng CYS, Wang KM, Kim TK, Jeayeng S, Slominski AT. CYP27A1 acts on the pre-vitamin D3 photoproduct, lumisterol, producing biologically active hydroxy-metabolites. J Steroid Biochem Mol Biol 2018; 181:1-10. [PMID: 29452159 PMCID: PMC5992068 DOI: 10.1016/j.jsbmb.2018.02.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/06/2018] [Accepted: 02/12/2018] [Indexed: 01/03/2023]
Abstract
Prolonged exposure of the skin to UV radiation causes previtamin D3, the initial photoproduct formed by opening of the B ring of 7-dehydrocholesterol, to undergo a second photochemical reaction where the B-ring is reformed giving lumisterol3 (L3), a stereoisomer of 7-dehydrocholesterol. L3 was believed to be an inactive photoproduct of excessive UV radiation whose formation prevents excessive vitamin D production. Recently, we reported that L3 is present in serum and that CYP11A1 can act on L3 producing monohydroxy- and dihydroxy-metabolites which inhibit skin cell proliferation similarly to 1α,25-dihydroxyvitamin D3. In this study we tested the ability of human CYP27A1 to hydroxylate L3. L3 was metabolized by purified CYP27A1 to 3 major products identified as 25-hydroxyL3, (25R)-27-hydroxyL3 and (25S)-27-hydroxyL3, by NMR. These three products were also seen when mouse liver mitochondria containing CYP27A1 were incubated with L3. The requirement for CYP27A1 for their formation by mitochondria was confirmed by the inhibition of their synthesis by 5β-cholestane-3α,7α,12α-triol, an intermediate in bile acid synthesis which serves as an efficient competitive substrate for CYP27A1. CYP27A1 displayed a high kcat for the metabolism of L3 (76 mol product/min/mol CYP27A1) and a catalytic efficiency (kcat/Km) that was 260-fold higher than that for vitamin D3. The CYP27A1-derived hydroxy-derivatives inhibited the proliferation of cultured human melanoma cells and colony formation with IC50 values in the nM range. Thus, L3 is metabolized efficiently by CYP27A1 with hydroxylation at C25 or C27 producing metabolites potent in their ability to inhibit melanoma cell proliferation, supporting that L3 is a prohormone which can be activated by CYP-dependent hydroxylations.
Collapse
Affiliation(s)
- Robert C Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia.
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dejian Ma
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Chloe Y S Cheng
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Katie M Wang
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Tae-Kang Kim
- Department of Dermatology, University of Alabama at Birmingham, AL 35294, USA
| | - Saowanee Jeayeng
- Department of Dermatology, University of Alabama at Birmingham, AL 35294, USA; Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, AL 35294, USA; VA Medical Center, Birmingham, AL 35294, USA
| |
Collapse
|
33
|
Karras SN, Koufakis T, Fakhoury H, Kotsa K. Deconvoluting the Biological Roles of Vitamin D-Binding Protein During Pregnancy: A Both Clinical and Theoretical Challenge. Front Endocrinol (Lausanne) 2018; 9:259. [PMID: 29875736 PMCID: PMC5974103 DOI: 10.3389/fendo.2018.00259] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/07/2018] [Indexed: 12/04/2022] Open
Abstract
The teleological purpose of an ongoing pregnancy is to fulfill its fundamental role of a successful, uncomplicated delivery, in conjunction with an optimal intrauterine environment for the developing fetus. Vitamin D metabolism is adapted to meet both these demands during pregnancy; first by stimulation of calcium absorption for adequate intrauterine bone mineral accrual of the fetus, and second, by enhancing systemic and local maternal tolerance to paternal and fetal alloantigens. Vitamin D-binding protein (VDBP) is one of the key biomolecules that optimize vitamin D homeostasis and also contributes as an immune regulator for a healthy, ongoing pregnancy. In this regard, recent results indicate that dysregulation of VDBP equilibrium could be a risk factor for adverse fetal, maternal, and neonatal outcomes, including preeclampsia, preterm birth, and gestational diabetes. Moreover, it has been hypothesized to be also implicated in the interpretation of vitamin D status in the pregnant state. The aim of this review is to assess available literature regarding the association of VDBP with clinical outcomes during pregnancy, as a potential biomarker for future clinical practice, with a discourse on current knowledge gaps and future research agenda.
Collapse
Affiliation(s)
- Spyridon N. Karras
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
- *Correspondence: Spyridon N. Karras,
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Hana Fakhoury
- Department of Biochemistry and Molecular Biology, College of Medicine, AlFaisal University, Riyadh, Saudi Arabia
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
34
|
Kao CY, Lin CN, Yang YL, Hamaguchi N, Yang SJ, Shen MC, Kao JT, Lin SW. Characterisation of factor IX with a glycine-to-valine missense mutation at residue 190 in a patient with severe haemophilia B. Thromb Haemost 2017; 105:616-26. [DOI: 10.1160/th10-11-0762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 01/06/2011] [Indexed: 11/05/2022]
Abstract
SummaryA patient with severe haemophilia B with a glycine-to-valine missense mutation at residue 190 (c25, chymotrypsin numbering) in factor IX (FIX; FIX-G190V or FIX-FuChou) had <1% of normal FIX clotting activity and 36% of normal FIX antigen levels (cross-reacting material-reduced, CRMr). Residue 190 in the C-terminal protease domain of human FIX is highly conserved in mammalian species and the serine protease family, suggesting that it has an indispensable role in protein function. To explore the pathological mechanism by which this mutation contributes to dysfunction of the FIX molecule, we functionally characterised FIX-G190V in vitro and in vivo. Liver-specific FIX-G190V gene expression following hydrodynamic plasmid delivery into haemophilia B mice revealed a 5.7-fold reduction in specific clotting activity compared with FIX-WT (wild type) and a two-fold decrease in plasma FIX-G190V concentration. Pulse-chase analysis demonstrated that FIX-G190V was secreted at a significantly slower rate than was FIX-WT. Purified FIX-G190V and FIX-WT displayed normal calcium-dependent conformational changes as shown by intrinsic fluorescence quenching. The in vivo half-lives of FIX-G190V and FIX-WT were indistinguishable. FIX-G190V was, however, more readily degraded than FIX-WT, especially after being activated by the active form of FXI. The vulnerable sites were mapped to the peptide bonds at Arg116-Leu117, Lys265-Tyr266, Arg327-Val328, and Arg338-Ser339, which are in the exposed loops of the FIX molecule. Also, failure of FXIa-activated FIX-G190V to bind p-aminobenzamidine indicated an abnormal conformation of the active-site pocket. Thus, the mutation at residue 190 of FIX may result in protein misfolding that affects secretion, clotting function, and hydrolysis.
Collapse
|
35
|
Mast N, Anderson KW, Lin JB, Li Y, Turko IV, Tatsuoka C, Bjorkhem I, Pikuleva IA. Cytochrome P450 27A1 Deficiency and Regional Differences in Brain Sterol Metabolism Cause Preferential Cholestanol Accumulation in the Cerebellum. J Biol Chem 2017; 292:4913-4924. [PMID: 28190002 DOI: 10.1074/jbc.m116.774760] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/10/2017] [Indexed: 01/04/2023] Open
Abstract
Cytochrome P450 27A1 (CYP27A1 or sterol 27-hydroxylase) is a ubiquitous, multifunctional enzyme catalyzing regio- and stereospecific hydroxylation of different sterols. In humans, complete CYP27A1 deficiency leads to cerebrotendinous xanthomatosis or nodule formation in tendons and brain (preferentially in the cerebellum) rich in cholesterol and cholestanol, the 5α-saturated analog of cholesterol. In Cyp27a1-/- mice, xanthomas are not formed, despite a significant cholestanol increase in the brain and cerebellum. The mechanism behind cholestanol production has been clarified, yet little is known about its metabolism, except that CYP27A1 might metabolize cholestanol. It also is unclear why CYP27A1 deficiency results in preferential cholestanol accumulation in the cerebellum. We hypothesized that cholestanol might be metabolized by CYP46A1, the principal cholesterol 24-hydroxylase in the brain. We quantified sterols along with CYP27A1 and CYP46A1 in mouse models (Cyp27a1-/-, Cyp46a1-/-, Cyp27a1-/-Cyp46a1-/-, and two wild type strains) and human brain specimens. In vitro experiments with purified P450s were conducted as well. We demonstrate that CYP46A1 is involved in cholestanol removal from the brain and that several factors contribute to the preferential increase in cholestanol in the cerebellum arising from CYP27A1 deficiency. These factors include (i) low cerebellar abundance of CYP46A1 and high cerebellar abundance of CYP27A1, the lack of which probably selectively increases the cerebellar cholestanol production; (ii) spatial separation in the cerebellum of cholesterol/cholestanol-metabolizing P450s from a pool of metabolically available cholestanol; and (iii) weak cerebellar regulation of cholesterol biosynthesis. We identified a new physiological role of CYP46A1, an important brain enzyme and cytochrome P450 that could be activated pharmacologically.
Collapse
Affiliation(s)
- Natalia Mast
- From the Departments of Ophthalmology and Visual Sciences and
| | - Kyle W Anderson
- the Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899.,the Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850, and
| | - Joseph B Lin
- From the Departments of Ophthalmology and Visual Sciences and
| | - Yong Li
- From the Departments of Ophthalmology and Visual Sciences and
| | - Illarion V Turko
- the Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899.,the Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850, and
| | - Curtis Tatsuoka
- Neurology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Ingemar Bjorkhem
- the Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, 141 86 Huddinge, Sweden
| | | |
Collapse
|
36
|
Mutemberezi V, Guillemot-Legris O, Muccioli GG. Oxysterols: From cholesterol metabolites to key mediators. Prog Lipid Res 2016; 64:152-169. [PMID: 27687912 DOI: 10.1016/j.plipres.2016.09.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/13/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Oxysterols are cholesterol metabolites that can be produced through enzymatic or radical processes. They constitute a large family of lipids (i.e. the oxysterome) involved in a plethora of physiological processes. They can act through GPCR (e.g. EBI2, SMO, CXCR2), nuclear receptors (LXR, ROR, ERα) and through transporters or regulatory proteins. Their physiological effects encompass cholesterol, lipid and glucose homeostasis. Additionally, they were shown to be involved in other processes such as immune regulatory functions and brain homeostasis. First studied as precursors of bile acids, they quickly emerged as interesting lipid mediators. Their levels are greatly altered in several pathologies and some oxysterols (e.g. 4β-hydroxycholesterol or 7α-hydroxycholestenone) are used as biomarkers of specific pathologies. In this review, we discuss the complex metabolism and molecular targets (including binding properties) of these bioactive lipids in human and mice. We also discuss the genetic mouse models currently available to interrogate their effects in pathophysiological settings. We also summarize the levels of oxysterols reported in two key organs in oxysterol metabolism (liver and brain), plasma and cerebrospinal fluid. Finally, we consider future opportunities and directions in the oxysterol field in order to gain a better insight and understanding of the complex oxysterol system.
Collapse
Affiliation(s)
- Valentin Mutemberezi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium.
| |
Collapse
|
37
|
Wang F, Yang L, Shi L, Li Q, Zhang G, Wu J, Zheng J, Jiao B. Nuclear translocation of fibroblast growth factor-2 (FGF2) is regulated by Karyopherin-β2 and Ran GTPase in human glioblastoma cells. Oncotarget 2016; 6:21468-78. [PMID: 26056081 PMCID: PMC4673279 DOI: 10.18632/oncotarget.4097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/13/2015] [Indexed: 11/25/2022] Open
Abstract
Human glioblastoma multiforme (GBM) is the most malignant tumor of the central nervous system (CNS). Fibroblast growth factor-2 (FGF2) belongs to the FGF superfamily and functions as a potential oncoprotein in GBM. FGF2 has low molecular weight (18K) and high molecular weight (HMW) isoforms. Nuclear accumulation of HMW-FGF2 strongly promotes glioblastoma cell proliferation, yet mechanism governing such cellular distribution remains unexplored. We investigated the mechanisms regulating FGF2 cellular localization in T98G human brain glioblastoma cells. We found HMW-FGF2, but not 18K-FGF2, is primarily located in the nucleus and interacts with nuclear transport protein Karyopherin-β2/Transportin (Kapβ2). SiRNA-directed Kapβ2 knockdown significantly reduced HMW-FGF2′s nuclear translocation. Moreover, inhibiting Ran GTPase activity also resulted in decreased HMW-FGF2 nuclear accumulation. Proliferation of T98G cells is greatly enhanced with transfections HMW-FGF2. Decreased PTEN expression and activated Akt signaling were observed upon HMW-FGF2 overexpression and might mediate pro-survival effect of FGF2. Interestingly, addition of nuclear localization signal (NLS) to 18K-FGF2 forced its nuclear import and dramatically increased cell proliferation and Akt activation. These findings demonstrated for the first time the molecular mechanisms for FGF2′s nuclear import, which promotes GBM cell proliferation and survival, providing novel insights to the development of GBM treatments.
Collapse
Affiliation(s)
- Feng Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Lijun Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Lin Shi
- Department of Neurosurgery, The Second Hospital of Baoding City, Baoding 071051, China
| | - Qian Li
- Department of Physiology, Hebei Medical University, Shijiazhuang 050000, China
| | - Gengshen Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Jianliang Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Jun Zheng
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Baohua Jiao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
38
|
Abstract
(25R)-26-Hydroxycholesterol (27-hydroxycholesterol) has been found to accumulate in breast tissue and to stimulate tumor growth via the estrogen receptor. Although most tissues express CYP27A1, the highest levels are in macrophages and most attention had been given to the production of 27-hydroxycholesterol in sub-endothelial macrophages as part of reverse cholesterol transport. In view of the newly identified biologic activity, it is important to consider the determinants of the levels of 27-hydroxycholesterol in macrophages that infiltrate breast tissue. Among these determinants are the oxysterol binding proteins expressed in macrophages, the level of expression of CYP7B1, the oxysterol 7 alpha hydroxylase that generates an inactive triol, and further oxidation of 27-hydroxycholestrol to the C27 acid by multifunctional CYP27A1. Transport of 27-hydroxycholesterol from macrophages to plasma is HDL-associated. In many tissues the ratio of 27-hydroxycholesterol to cholesterol (ng/μg) is higher than that in plasma. Tamoxifen, an effective estrogen receptor antagonist that prevents breast cancer, also has the biologic property of blocking several steps in the lanosterol to cholesterol metabolic pathway. In genetically disposed women, tamoxifen may increase the amount of 27-hydroxycholesterol in breast tissue.
Collapse
|
39
|
Bandaru VVR, Haughey NJ. Quantitative detection of free 24S-hydroxycholesterol, and 27-hydroxycholesterol from human serum. BMC Neurosci 2014; 15:137. [PMID: 25539717 PMCID: PMC4304132 DOI: 10.1186/s12868-014-0137-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 12/17/2014] [Indexed: 03/25/2023] Open
Abstract
Background Cholesterol metabolism is important for the maintenance of myelin and neuronal membranes in the central nervous system. Blood concentrations of the brain specific cholesterol metabolite 24S-hydroxysterol to the peripheral metabolite 27-hydroxycholesterol may be useful surrogate markers for neurodegenerative diseases including Alzheimer’s disease, Huntington’s disease, HIV-Associated Neurocognitive Disorders, and Multiple Sclerosis. However, current methods to isolate hydroxycholesterols are labor intensive, prone to produce variable extraction efficiencies and do not discriminate between free and esterfied forms of hydroxycholesterols. Since free hydroxycholesterols are the biologically active form of these sterols, separating free from esterfied forms may provide a sensitive measure to identify disease-associated differences in brain sterol metabolism. Results We found that average human serum concentrations were 12.3 ± 4.79 ng/ml for free 24(s)-hydroxycholesterol and 17.7 ± 8.5 ng/ml for 27-hydroxycholesterol. Conclusion Serum measurements of these biologically active oxysterols may be useful surrogate measures for brain health in a variety of neurodegenerative conditions.
Collapse
Affiliation(s)
- Veera Venkata Ratnam Bandaru
- Department of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections, The Johns Hopkins University School of Medicine, Carnegie 616A, 600 North Wolfe Street, Baltimore, 21287, MD, USA.
| | - Norman J Haughey
- Department of Neurology, Richard T. Johnson Division of Neuroimmunology and Neurological Infections, The Johns Hopkins University School of Medicine, Carnegie 616A, 600 North Wolfe Street, Baltimore, 21287, MD, USA. .,Department of Psychiatry, Division of Geriatric Psychiatry and Neuropsychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
40
|
Lee WR, Ishikawa T, Umetani M. The interaction between metabolism, cancer and cardiovascular disease, connected by 27-hydroxycholesterol. ACTA ACUST UNITED AC 2014; 9:617-624. [PMID: 25632306 DOI: 10.2217/clp.14.53] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oxysterols are metabolites of cholesterol that are produced in liver and other peripheral tissues as a means to eliminate cholesterol to bile acid. Recent studies have revealed that the most abundant circulating oxysterol 27-hydroxycholesterol (27HC) is the first identified endogenous selective estrogen receptor modulator. 27HC levels correlate well with that of cholesterol, and also rise progressively with age. 27HC affects estrogen receptor function by the antagonism of estrogen action and also by the direct modulation of the receptor function, and similar to estrogen/estrogen receptors, 27HC has many actions in various tissues. This review article introduces the recent progress in the understanding of the role of 27HC in breast cancer and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Wan-Ru Lee
- Division of Pulmonary & Vascular Biology, Departments of Pediatrics & Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Tomonori Ishikawa
- Division of Pulmonary & Vascular Biology, Departments of Pediatrics & Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA ; Comprehensive Reproductive Medicine, Graduate School of Medical & Dental Sciences, Tokyo Medical & Dental University, Tokyo, Japan
| | - Michihisa Umetani
- Division of Pulmonary & Vascular Biology, Departments of Pediatrics & Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
41
|
Bonny O, Bochud M. Genetics of calcium homeostasis in humans: continuum between monogenic diseases and continuous phenotypes. Nephrol Dial Transplant 2014; 29 Suppl 4:iv55-62. [PMID: 25165186 DOI: 10.1093/ndt/gfu195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Extracellular calcium participates in several key physiological functions, such as control of blood coagulation, bone calcification or muscle contraction. Calcium homeostasis in humans is regulated in part by genetic factors, as illustrated by rare monogenic diseases characterized by hypo or hypercalcaemia. Both serum calcium and urinary calcium excretion are heritable continuous traits in humans. Serum calcium levels are tightly regulated by two main hormonal systems, i.e. parathyroid hormone and vitamin D, which are themselves also influenced by genetic factors. Recent technological advances in molecular biology allow for the screening of the human genome at an unprecedented level of detail and using hypothesis-free approaches, such as genome-wide association studies (GWAS). GWAS identified novel loci for calcium-related phenotypes (i.e. serum calcium and 25-OH vitamin D) that shed new light on the biology of calcium in humans. The substantial overlap (i.e. CYP24A1, CASR, GATA3; CYP2R1) between genes involved in rare monogenic diseases and genes located within loci identified in GWAS suggests a genetic and phenotypic continuum between monogenic diseases of calcium homeostasis and slight disturbances of calcium homeostasis in the general population. Future studies using whole-exome and whole-genome sequencing will further advance our understanding of the genetic architecture of calcium homeostasis in humans. These findings will likely provide new insight into the complex mechanisms involved in calcium homeostasis and hopefully lead to novel preventive and therapeutic approaches. Keyword: calcium, monogenic, genome-wide association studies, genetics.
Collapse
Affiliation(s)
- Olivier Bonny
- Service of Nephrology, Lausanne University Hospital, Lausanne, Switzerland Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Murielle Bochud
- Community Prevention Unit, Institute for Social and Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
42
|
Barau C, Ghaleh B, Berdeaux A, Morin D. Cytochrome P450 and myocardial ischemia: potential pharmacological implication for cardioprotection. Fundam Clin Pharmacol 2014; 29:1-9. [DOI: 10.1111/fcp.12087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/20/2014] [Accepted: 06/13/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Caroline Barau
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| | - Bijan Ghaleh
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| | - Alain Berdeaux
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| | - Didier Morin
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| |
Collapse
|
43
|
Boyd M, Coskun M, Lilje B, Andersson R, Hoof I, Bornholdt J, Dahlgaard K, Olsen J, Vitezic M, Bjerrum JT, Seidelin JB, Nielsen OH, Troelsen JT, Sandelin A. Identification of TNF-α-responsive promoters and enhancers in the intestinal epithelial cell model Caco-2. DNA Res 2014; 21:569-83. [PMID: 24990076 PMCID: PMC4263293 DOI: 10.1093/dnares/dsu022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Caco-2 cell line is one of the most important in vitro models for enterocytes, and is used to study drug absorption and disease, including inflammatory bowel disease and cancer. In order to use the model optimally, it is necessary to map its functional entities. In this study, we have generated genome-wide maps of active transcription start sites (TSSs), and active enhancers in Caco-2 cells with or without tumour necrosis factor (TNF)-α stimulation to mimic an inflammatory state. We found 520 promoters that significantly changed their usage level upon TNF-α stimulation; of these, 52% are not annotated. A subset of these has the potential to confer change in protein function due to protein domain exclusion. Moreover, we locate 890 transcribed enhancer candidates, where ∼50% are changing in usage after TNF-α stimulation. These enhancers share motif enrichments with similarly responding gene promoters. As a case example, we characterize an enhancer regulating the laminin-5 γ2-chain (LAMC2) gene by nuclear factor (NF)-κB binding. This report is the first to present comprehensive TSS and enhancer maps over Caco-2 cells, and highlights many novel inflammation-specific promoters and enhancers.
Collapse
Affiliation(s)
- Mette Boyd
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark
| | - Mehmet Coskun
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark Department of Gastroenterology, Medical Section, University of Copenhagen, Herlev Hospital, Herlev DK-2730, Denmark
| | - Berit Lilje
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark
| | - Robin Andersson
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark
| | - Ilka Hoof
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark
| | - Jette Bornholdt
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark
| | - Katja Dahlgaard
- Department of Science, Systems and Models, Roskilde University, Roskilde DK-4000, Denmark
| | - Jørgen Olsen
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Morana Vitezic
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark
| | - Jacob Tveiten Bjerrum
- Department of Gastroenterology, Medical Section, University of Copenhagen, Herlev Hospital, Herlev DK-2730, Denmark
| | - Jakob Benedict Seidelin
- Department of Gastroenterology, Medical Section, University of Copenhagen, Herlev Hospital, Herlev DK-2730, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Medical Section, University of Copenhagen, Herlev Hospital, Herlev DK-2730, Denmark
| | | | - Albin Sandelin
- The Bioinformatics Centre, Department of Biology & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloes Vej 5, Copenhagen DK-2200, Denmark
| |
Collapse
|
44
|
Cholesterol in the retina: the best is yet to come. Prog Retin Eye Res 2014; 41:64-89. [PMID: 24704580 DOI: 10.1016/j.preteyeres.2014.03.002] [Citation(s) in RCA: 205] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 03/13/2014] [Accepted: 03/17/2014] [Indexed: 01/09/2023]
Abstract
Historically understudied, cholesterol in the retina is receiving more attention now because of genetic studies showing that several cholesterol-related genes are risk factors for age-related macular degeneration (AMD) and because of eye pathology studies showing high cholesterol content of drusen, aging Bruch's membrane, and newly found subretinal lesions. The challenge before us is determining how the cholesterol-AMD link is realized. Meeting this challenge will require an excellent understanding these genes' roles in retinal physiology and how chorioretinal cholesterol is maintained. In the first half of this review, we will succinctly summarize physico-chemical properties of cholesterol, its distribution in the human body, general principles of maintenance and metabolism, and differences in cholesterol handling in human and mouse that impact on experimental approaches. This information will provide a backdrop to the second part of the review focusing on unique aspects of chorioretinal cholesterol homeostasis, aging in Bruch's membrane, cholesterol in AMD lesions, a model for lesion biogenesis, a model for macular vulnerability based on vascular biology, and alignment of AMD-related genes and pathobiology using cholesterol and an atherosclerosis-like progression as unifying features. We conclude with recommendations for the most important research steps we can take towards delineating the cholesterol-AMD link.
Collapse
|
45
|
Ishikawa T, Wakabayashi-Nakao K, Nakagawa H. Methods to examine the impact of nonsynonymous SNPs on protein degradation and function of human ABC transporter. Methods Mol Biol 2014; 1015:225-50. [PMID: 23824860 DOI: 10.1007/978-1-62703-435-7_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Clinical studies have strongly suggested that genetic polymorphisms and/or mutations of certain ATP-binding cassette (ABC) transporter genes might be regarded as significant factors affecting patients' responses to medication and/or the risk of diseases. In the case of ABCG2, certain single nucleotide polymorphisms (SNPs) in the encoding gene alter the substrate specificity and/or enhance endoplasmic reticulum-associated degradation (ERAD) of the de novo synthesized ABCG2 protein via the ubiquitin-mediated proteasomal proteolysis pathway. Hitherto accumulated clinical data imply that several nonsynonymous SNPs affect the ABCG2-mediated clearance of drugs or cellular metabolites, although some controversies still exist. Therefore, we recently developed high-speed functional screening and ERAD of ABC transporters so as to evaluate the effect of genetic polymorphisms on their function and protein expression levels in vitro. In this chapter we present in vitro experimental methods to elucidate the impact of nonsynonymous SNPs on protein degradation of ABCG2 as well as on its transport function.
Collapse
|
46
|
Coordinated Transcriptional Regulation of Hspa1a Gene by Multiple Transcription Factors: Crucial Roles for HSF-1, NF-Y, NF-κB, and CREB. J Mol Biol 2014; 426:116-35. [DOI: 10.1016/j.jmb.2013.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/09/2013] [Accepted: 09/09/2013] [Indexed: 11/20/2022]
|
47
|
Sun N, Bao Z, Xiong X, Zhao H. SunnyTALEN: A second-generation TALEN system for human genome editing. Biotechnol Bioeng 2013; 111:683-91. [DOI: 10.1002/bit.25154] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 10/17/2013] [Accepted: 11/14/2013] [Indexed: 12/18/2022]
Affiliation(s)
- Ning Sun
- Department of Biochemistry; University of Illinois at Urbana-Champaign; 600 South Mathews Avenue Urbana Illinois 61801
| | - Zehua Bao
- Department of Biochemistry; University of Illinois at Urbana-Champaign; 600 South Mathews Avenue Urbana Illinois 61801
| | - Xiong Xiong
- Department of Chemical and Biomolecular Engineering; University of Illinois at Urbana-Champaign; 600 South Mathews Avenue Urbana Illinois 61801
| | - Huimin Zhao
- Department of Biochemistry; University of Illinois at Urbana-Champaign; 600 South Mathews Avenue Urbana Illinois 61801
- Department of Chemical and Biomolecular Engineering; University of Illinois at Urbana-Champaign; 600 South Mathews Avenue Urbana Illinois 61801
- Department of Bioengineering, Department of Chemistry; Center for Biophysics and Computational Biology and Institute for Genomic Biology; University of Illinois at Urbana-Champaign; 600 South Mathews Avenue Urbana Illinois 61801
| |
Collapse
|
48
|
Cherry AL, Finta C, Karlström M, Jin Q, Schwend T, Astorga-Wells J, Zubarev RA, Del Campo M, Criswell AR, de Sanctis D, Jovine L, Toftgård R. Structural basis of SUFU-GLI interaction in human Hedgehog signalling regulation. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:2563-79. [PMID: 24311597 PMCID: PMC3852661 DOI: 10.1107/s0907444913028473] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 10/16/2013] [Indexed: 12/13/2022]
Abstract
Hedgehog signalling plays a fundamental role in the control of metazoan development, cell proliferation and differentiation, as highlighted by the fact that its deregulation is associated with the development of many human tumours. SUFU is an essential intracellular negative regulator of mammalian Hedgehog signalling and acts by binding and modulating the activity of GLI transcription factors. Despite its central importance, little is known about SUFU regulation and the nature of SUFU-GLI interaction. Here, the crystal and small-angle X-ray scattering structures of full-length human SUFU and its complex with the key SYGHL motif conserved in all GLIs are reported. It is demonstrated that GLI binding is associated with major conformational changes in SUFU, including an intrinsically disordered loop that is also crucial for pathway activation. These findings reveal the structure of the SUFU-GLI interface and suggest a mechanism for an essential regulatory step in Hedgehog signalling, offering possibilities for the development of novel pathway modulators and therapeutics.
Collapse
Affiliation(s)
- Amy L Cherry
- Department of Biosciences and Nutrition and Center for Biosciences, Karolinska Institutet, Novum, Hälsovägen 7, SE-141 83 Huddinge, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gabbi C, Warner M, Gustafsson JÅ. Action mechanisms of Liver X Receptors. Biochem Biophys Res Commun 2013; 446:647-50. [PMID: 24300092 DOI: 10.1016/j.bbrc.2013.11.077] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 11/19/2013] [Indexed: 12/18/2022]
Abstract
The two Liver X Receptors, LXRα and LXRβ, are nuclear receptors belonging to the superfamily of ligand-activated transcription factors. They share more than 78% homology in amino acid sequence, a common profile of oxysterol ligands and the same heterodimerization partner, Retinoid X Receptor. LXRs play crucial roles in several metabolic pathways: lipid metabolism, in particular in preventing cellular cholesterol accumulation; glucose homeostasis; inflammation; central nervous system functions and water transport. As with all nuclear receptors, the transcriptional activity of LXR is the result of an orchestration of numerous cellular factors including ligand bioavailability, presence of corepressors and coactivators and cellular context i.e., what other pathways are activated in the cell at the time the receptor recognizes its ligand. In this mini-review we summarize the factors regulating the transcriptional activity and the mechanisms of action of these two receptors.
Collapse
Affiliation(s)
- Chiara Gabbi
- Center for Nuclear Receptors and Cell Signaling, University of Houston, 3056 Cullen Blv, 77204 Houston, Texas, USA
| | - Margaret Warner
- Center for Nuclear Receptors and Cell Signaling, University of Houston, 3056 Cullen Blv, 77204 Houston, Texas, USA
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, University of Houston, 3056 Cullen Blv, 77204 Houston, Texas, USA; Department of Biosciences and Nutrition, Karolinska Institutet, Novum S-141 86, Sweden.
| |
Collapse
|
50
|
Kang GY, Pyun BJ, Seo HR, Jin YB, Lee HJ, Lee YJ, Lee YS. Inhibition of Snail1-DNA-PKcs protein-protein interface sensitizes cancer cells and inhibits tumor metastasis. J Biol Chem 2013; 288:32506-32516. [PMID: 24085291 DOI: 10.1074/jbc.m113.479840] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our previous study suggested that the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) interacts with Snail1, which affects genomic instability, sensitivity to DNA-damaging agents, and migration of tumor cells by reciprocal regulation between DNA-PKcs and Snail1. Here, we further investigate that a peptide containing 7-amino acid sequences (amino acids 15-21) of Snail1 (KPNYSEL, SP) inhibits the endogenous interaction between DNA-PKcs and Snail1 through primary interaction with DNA-PKcs. SP restored the inhibited DNA-PKcs repair activity and downstream pathways. On the other hand, DNA-PKcs-mediated phosphorylation of Snail1 was inhibited by SP, which resulted in decreased Snail1 stability and Snail1 functions. However, these phenomena were only shown in p53 wild-type cells, not in p53-defective cells. From these results, it is suggested that interfering with the protein interaction between DNA-PKcs and Snail1 might be an effective strategy for sensitizing cancer cells and inhibiting tumor migration, especially in both Snail1-overexpressing and DNA-PKcs-overexpressing cancer cells with functional p53.
Collapse
Affiliation(s)
- Ga-Young Kang
- From the College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750
| | - Bo-Jeong Pyun
- From the College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750
| | - Haeng Ran Seo
- the Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706
| | - Yeung Bae Jin
- the Korea Atomic Energy Research Institute, Advanced Radiation Technology Institute, Jeongeup-si, Jeollabuk-do 580-185, Korea
| | - Hae-June Lee
- the Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706
| | - Yoon-Jin Lee
- the Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul 139-706
| | - Yun-Sil Lee
- From the College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750,.
| |
Collapse
|