1
|
Frungieri MB, Calandra RS, Matzkin ME, Rossi SP. Melatonin as a natural anti-inflammatory and anti-oxidant therapy in the testis: a focus on infertility and aging†. Biol Reprod 2024; 111:543-556. [PMID: 38869910 DOI: 10.1093/biolre/ioae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Melatonin is a pineal hormone that regulates testicular activity (i.e., steroidogenesis and spermatogenesis) through two complementary mechanisms, indirect effects exerted via the hypothalamic-adenohypophyseal axis and direct actions that take place on the different cell populations of the male gonad. The effects of increased age on the testis and the general mechanisms involved in testicular pathology leading to infertility are still only poorly understood. However, there is growing evidence that link testicular aging and idiopathic male infertility to local inflammatory and oxidative stress events. Because literature data strongly indicate that melatonin exhibits anti-inflammatory and anti-oxidant properties, this review focuses on the potential benefits exerted by this indoleamine at testicular level in male reproductive fertility and aging. Taking into account that the effects of melatonin supplementation on testicular function are currently being investigated, the overview covers not only promising prospects but also many questions concerning the future therapeutic value of this indoleamine as an anti-aging drug as well as in the management of cases of male infertility for which there are no medical treatments currently available.
Collapse
Affiliation(s)
- Mónica Beatriz Frungieri
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
| | - Ricardo Saúl Calandra
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
| | - María Eugenia Matzkin
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
- Cátedra 1, Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Soledad Paola Rossi
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
- Cátedra 1, Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
2
|
Kim NH, Lee SJ, Lee KJ, Song AR, Park HJ, Kang JS, Cha JY, Han YH. The Root Extract of Rosa multiflora Ameliorates Nonalcoholic Steatohepatitis Development via Blockade of De Novo Lipogenesis and Inflammation. Curr Issues Mol Biol 2024; 46:5881-5893. [PMID: 38921022 PMCID: PMC11202599 DOI: 10.3390/cimb46060351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is characterized by severe inflammation and fibrosis due to an excessive accumulation of triglycerides (TGs) in the liver with a dysregulated de novo lipogenesis (DNL) pathway. In this study, we aimed to evaluate the effectiveness of YC-1102, an extract obtained from the roots of Rosa multiflora, as a nutritional supplement in a diet-induced NASH mouse model. C57BL/6 wild-type mice were fed a fructose, palmitate, and cholesterol (FPC)-containing diet for 16 weeks to induce experimental NASH. A daily oral gavage of YC-1102 and obetichoic acid (OCA) was conducted for 9 weeks. After sacrifice, disease parameters related to hepatic lipids, inflammation, and fibrosis were evaluated. The treatment with YC-1102 significantly decreased the liver/body weight ratio, epididymal fat weight, and plasma ALT and AST levels, which are indicators of NASH injuries. YC-1102 attenuated hepatic lipid accumulation by inhibiting the transcription of DNL genes in the livers exhibiting NASH. Additionally, we found that YC-1102 blocked the development of hepatic inflammation and fibrosis by directly disturbing macrophage activation, resulting in an amelioration of hepatic fibrosis. Our findings suggest that YC-1102 could ameliorate NASH progression by inhibiting uncontrolled DNL and inflammation.
Collapse
Affiliation(s)
- Nam-Hee Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
| | - Seung-Jin Lee
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
| | - Kyeong-Jin Lee
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
| | - Ae Ri Song
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Hyun-Je Park
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Jong Soo Kang
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Joo Young Cha
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea; (A.R.S.); (H.-J.P.); (J.S.K.)
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.-H.K.); (S.-J.L.); (K.-J.L.)
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
3
|
Ford HR, Bionaz M. The Experimental and In Silico-Based Evaluation of NRF2 Modulators, Sulforaphane and Brusatol, on the Transcriptome of Immortalized Bovine Mammary Alveolar Cells. Int J Mol Sci 2024; 25:4264. [PMID: 38673850 PMCID: PMC11049820 DOI: 10.3390/ijms25084264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Changes during the production cycle of dairy cattle can leave these animals susceptible to oxidative stress and reduced antioxidant health. In particular, the periparturient period, when dairy cows must rapidly adapt to the sudden metabolic demands of lactation, is a period when the production of damaging free radicals can overwhelm the natural antioxidant systems, potentially leading to tissue damage and reduced milk production. Central to the protection against free radical damage and antioxidant defense is the transcription factor NRF2, which activates an array of genes associated with antioxidant functions and cell survival. The objective of this study was to evaluate the effect that two natural NRF2 modulators, the NRF2 agonist sulforaphane (SFN) and the antagonist brusatol (BRU), have on the transcriptome of immortalized bovine mammary alveolar cells (MACT) using both the RT-qPCR of putative NRF2 target genes, as well as RNA sequencing approaches. The treatment of cells with SFN resulted in the activation of many putative NRF2 target genes and the upregulation of genes associated with pathways involved in cell survival, metabolism, and antioxidant function while suppressing the expression of genes related to cellular senescence and DNA repair. In contrast, the treatment of cells with BRU resulted in the upregulation of genes associated with inflammation, cellular stress, and apoptosis while suppressing the transcription of genes involved in various metabolic processes. The analysis also revealed several novel putative NRF2 target genes in bovine. In conclusion, these data indicate that the treatment of cells with SFN and BRU may be effective at modulating the NRF2 transcriptional network, but additional effects associated with cellular stress and metabolism may complicate the effectiveness of these compounds to improve antioxidant health in dairy cattle via nutrigenomic approaches.
Collapse
Affiliation(s)
| | - Massimo Bionaz
- Department of Animal and Rangeland Science, Oregon State University, Corvallis, OR 97331, USA;
| |
Collapse
|
4
|
Kim H, Lee S, Jeong C, Han Y, Lee M. RORα-GABP-TFAM axis alleviates myosteatosis with fatty atrophy through reinforcement of mitochondrial capacity. J Cachexia Sarcopenia Muscle 2024; 15:615-630. [PMID: 38272857 PMCID: PMC10995264 DOI: 10.1002/jcsm.13432] [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: 06/11/2023] [Revised: 11/22/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Fat infiltration in muscle, called 'myosteatosis', precedes muscle atrophy, which subsequently results in sarcopenia. Myosteatosis is frequently observed in patients with nonalcoholic fatty liver disease (NAFLD). We have previously reported that retinoic acid receptor-related orphan receptor-α (RORα) regulates mitochondrial dynamics and mitophagy in hepatocytes, resulting in an alleviation of NAFLD. In this study, we aimed to investigate the role of RORα in skeletal muscle and to understand molecular mechanisms by which RORα controls mitochondrial capacity, using an NAFLD-associated myosteatosis mouse model. METHODS To establish a myosteatosis model, 7-week-old C57BL/6N mice were fed with high-fat diet (HFD). After 15 weeks of diet feeding, an adeno-associated virus vector encoding RORα (AAV-RORα) was injected to gastrocnemius (GA) muscles, or after 7 weeks of HFD feeding, JC1-40, an RORα agonistic ligand, was administered daily at a dose of 5 mg/kg/day by oral gavage for 5 weeks. Histological, biochemical and molecular analyses in various in vivo and in vitro experiments were performed. RESULTS First, the number of oxidative MyHC2a fibres with intensive lipid infiltration increased by 3.8-fold in the red region of the GA of mice with myosteatosis (P < 0.001). RORα was expressed around MyHC2a fibres, and its level increased by 2.7-fold after HFD feeding (P < 0.01). Second, treatment of RORα ligands in C2C12 myoblasts, such as cholesterol sulfate and JC1-40, enhanced the number of oxidative fibres stained for MyHC1 and MyHC2a by two-fold to four-fold (P < 0.01), while it reduced the lipid levels in MyHC2a fibres by 20-50% (P < 0.001) in the presence of palmitic acids. Third, mitochondrial membrane potential (P < 0.01) and total area of mitochondria (P < 0.01) were enhanced by treatment of these ligands. Chromatin immunoprecipitation analysis showed that RORα bound the promoter of GA-binding protein α subunit gene that led to activation of mitochondrial transcription factor A (TFAM) in C2C12 myoblasts (P < 0.05). Finally, intramuscular transduction of AAV-RORα alleviated the HFD-induced myosteatosis with fatty atrophy; lipid contents in MyHC2a fibres decreased by 48% (P < 0.001), whereas the number of MyHC2b fibre increased by 22% (P < 0.001). Also, administration of JC1-40 improved the signs of myosteatosis in that it decreased the level of adipose differentiation-related protein (P < 0.01) but increased mitochondrial proteins such as cytochrome c oxidase 4 and TFAM in GA muscle (P < 0.01). CONCLUSIONS RORα plays a versatile role in regulating the quantity of mitochondria and the oxidative capacity, ultimately leading to an improvement in myosteatosis symptoms.
Collapse
Affiliation(s)
- Hyeon‐Ji Kim
- College of PharmacySeoul National UniversitySeoulSouth Korea
- Research Institute of Pharmaceutical SciencesSeoulSouth Korea
| | - Sang‐Heon Lee
- College of PharmacySeoul National UniversitySeoulSouth Korea
| | - Cheolhee Jeong
- College of PharmacySeoul National UniversitySeoulSouth Korea
| | - Yong‐Hyun Han
- College of PharmacyKangwon National UniversityChuncheonSouth Korea
| | - Mi‐Ock Lee
- College of PharmacySeoul National UniversitySeoulSouth Korea
- Research Institute of Pharmaceutical SciencesSeoulSouth Korea
- Bio‐MAX InstituteSeoul National UniversitySeoulSouth Korea
| |
Collapse
|
5
|
Choi H, Oh D, Kim HJ, Chambugong M, Kim MH, Lee MO, Park HG. An RORα agonist, ODH-08, inhibits fibrogenic activation of hepatic stellate cells via suppression of SMAD3. Life Sci 2024; 340:122443. [PMID: 38242496 DOI: 10.1016/j.lfs.2024.122443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/19/2023] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
AIMS Hepatic fibrosis is a dynamic process characterized by the net accumulation of an extracellular matrix resulting from chronic liver injury such as nonalcoholic steatohepatitis. Activation of hepatic stellate cells (HSCs) plays a role in transdifferentiation of quiescent cells into fibrogenic myofibroblasts. We aimed to examine the function of retinoic acid receptor-related orphan receptor alpha (RORα) and its novel agonistic ligand, 1-(4-benzyloxybenzyl)-3-(2-dimethylaminoethyl)-thiourea (ODH-08) against activation of HSCs using hepatic fibrosis mouse models. MAIN METHODS Chemical synthesis, a reporter gene assay, surface plasmon resonance analysis, and a docking study were performed to evaluate ODH-08 as a ligand of RORα. In vivo experiments with mice fed a Western diet were performed to evaluate the effect of ODH-08. The human HSC line, Lx-2, and primary mouse HSCs were employed to identify the molecular mechanisms underlying the antifibrogenic effect of ODH-08. KEY FINDINGS A novel RORα-selective ligand, ODH-08, was developed based on modification of JC1-40, an analog of N-methylthiourea. Administration of ODH-08 to the Western diet-fed mice reduced hepatic collagen deposition and expression levels of fibrogenic markers such as α-smooth muscle actin and collagen type I alpha 1 chain. Activation of RORα-either by transient overexpression of RORα or treatment with ODH-08-suppressed the expression of fibrogenic proteins in HSCs. The activation of RORα suppressed the activity of SMAD2 and 3, which are the primary downstream proteins of transforming growth factor β. SIGNIFICANCE RORα and its agonist ODH-08 have a potent antifibrotic effect, which could provide a novel antifibrotic strategy against hepatic fibrosis.
Collapse
Affiliation(s)
- Haena Choi
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Daehyun Oh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul, Republic of Korea
| | - Melody Chambugong
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Mi-Hyun Kim
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul, Republic of Korea; Bio-MAX Institute, Seoul National University, Seoul, Republic of Korea.
| | - Hyeung-Geun Park
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Kim MY, Jeong B, Lee GS, Jeon H, Yang YM, Yang H, Han YH. Panaxydol extracted from Panax ginseng inhibits NLRP3 inflammasome activation to ameliorate NASH-induced liver injury. Int Immunopharmacol 2024; 128:111565. [PMID: 38262161 DOI: 10.1016/j.intimp.2024.111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/25/2024]
Abstract
Activation of NOD-like receptor protein 3 (NLRP3) inflammasome exacerbates liver inflammation and fibrosis in nonalcoholic steatohepatitis (NASH), suggesting that development of inflammasome inhibitor can become leading candidate to ameliorate NASH. Panax ginseng (P. ginseng) contains numerous bioactive natural components to reduce inflammation. This study aims to identify inhibitory components of P. ginseng for NLRP3 inflammasome activation. We separated polar and non-polar fractions of P. ginseng and tested modulation of NLRP3 inflammasome, and then identified pure component for inflammasome inhibitor which ameliorates diet-induced NASH. Non-polar P. ginseng fractions obtained from ethyl acetate solvent attenuated IL-1β secretion and expression of active caspase-1. We revealed that panaxydol (PND) is pure component to inhibit NLRP3 inflammasome activation. PND blocked inflammasome cytokines release, pyroptotic cell death, caspase-1 activation and specking of inflammasome complex. Inhibitory effect of PND was specific to NLRP3-dependent pathway via potential interaction with ATP binding motif of NLRP3. Moreover, in vivo studies showed that PND plays beneficial roles to reduce tissue inflammations through disruption of NLRP3 inflammasome and to ameliorate the development of NASH. These results provide new insight of natural products, panaxydol, for NLRP3 inflammasome inhibitor and could offer potential therapeutic candidate for reliving NASH.
Collapse
Affiliation(s)
- Mi-Yeon Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Birang Jeong
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, South Korea
| | - Hongjun Jeon
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Yoon Mee Yang
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea; College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Heejung Yang
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea.
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea; Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea.
| |
Collapse
|
7
|
Shen Y, Tang Q, Wang J, Zhou Z, Yin Y, Zhang Y, Zheng W, Wang X, Chen G, Sun J, Chen L. Targeting RORα in macrophages to boost diabetic bone regeneration. Cell Prolif 2023; 56:e13474. [PMID: 37051760 PMCID: PMC10542986 DOI: 10.1111/cpr.13474] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Diabetes mellitus (DM) has become a serious threat to human health. Bone regeneration deficiency and nonunion caused by DM is perceived as a worldwide epidemic, with a very high socioeconomic impact on public health. Here, we find that targeted activation of retinoic acid-related orphan receptor α (RORα) by SR1078 in the early stage of bone defect repair can significantly promote in situ bone regeneration of DM rats. Bone regeneration relies on the activation of macrophage RORα in the early bone repair, but RORα of DM rats fails to upregulation as hyperglycemic inflammatory microenvironment induced IGF1-AMPK signalling deficiency. Mechanistic investigations suggest that RORα is vital for macrophage-induced migration and proliferation of bone mesenchymal stem cells (BMSCs) via a CCL3/IL-6 depending manner. In summary, our study identifies RORα expressed in macrophages during the early stage of bone defect repair is crucial for in situ bone regeneration, and offers a novel strategy for bone regeneration therapy and fracture repair in DM patients.
Collapse
Affiliation(s)
- Yufeng Shen
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
- Department of Stomatology, The First Affiliated Hospital, School of MedicineShihezi UniversityShihezi 832000China
| | - Qingming Tang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Jiajia Wang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Zheng Zhou
- Department of Stomatology, The First Affiliated Hospital, School of MedicineShihezi UniversityShihezi 832000China
| | - Ying Yin
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Yifan Zhang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Wenhao Zheng
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Xinyuan Wang
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Guangjin Chen
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Jiwei Sun
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| | - Lili Chen
- Department of StomatologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022China
- School of StomatologyTongji Medical College, Huazhong University of Science and TechnologyWuhan 430030China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhan 430022China
| |
Collapse
|
8
|
Zhang S, Shen J, Zhu Y, Zheng Y, San W, Cao D, Chen Y, Meng G. Hydrogen sulfide promoted retinoic acid-related orphan receptor α transcription to alleviate diabetic cardiomyopathy. Biochem Pharmacol 2023; 215:115748. [PMID: 37591449 DOI: 10.1016/j.bcp.2023.115748] [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: 05/16/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Diabetic cardiomyopathy (DCM) is one serious and common complication in diabetes without effective treatments. Hydrogen sulfide (H2S) fights against a variety of cardiovascular diseases including DCM. Retinoic acid-related orphan receptor α (RORα) has protective effects on cardiovascular system. However, whether RORα mediates the protective effect of H2S against DCM remains unknown. The present research was to explore the roles and mechanisms of RORα in H2S against DCM. The study demonstrated that H2S donor sodium hydrosulfide (NaHS) alleviated cell injury but enhanced RORα expression in high glucose (HG)-stimulated cardiomyocytes. However, NaHS no longer had the protective effect on attenuating cell damage and oxidative stress, improving mitochondrial membrane potential, inhibiting necroptosis and enhanced signal transducer and activator of transcription 3 (STAT3) Ser727 phosphorylation in HG-stimulated cardiomyocytes after RORα siRNA transfection. Moreover, NaHS improved cardiac function, attenuated myocardial hypertrophy and fibrosis, alleviated oxidative stress, inhibited necroptosis, but increased STAT3 phosphorylation in wild type (WT) mice but not in RORα knockout mice (a spontaneous staggerer mice, sg/sg mice) with diabetes. Additionally, NaHS increased RORα promoter activity in cardiomyocytes with HG stimulation, which was related to the binding sites of E2F transcription factor 1 (E2F1) in the upstream region of RORα promoter. NaHS enhanced E2F1 expression and increased the binding of E2F1 to RORα promoter in cardiomyocytes with HG stimulation. In sum, H2S promoted RORα transcription via E2F1 to alleviate necroptosis and protect against DCM. It is helpful to propose a novel therapeutic implication for DCM.
Collapse
Affiliation(s)
- Shuping Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China; Department of Pharmacy, Nantong Third People's Hospital; Department of Pharmacy, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226001, Jiangsu, China
| | - Jieru Shen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Yu Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Yangyang Zheng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Wenqing San
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Danyi Cao
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Yun Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China.
| | - Guoliang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
9
|
Karri K, Waxman DJ. TCDD dysregulation of lncRNA expression, liver zonation and intercellular communication across the liver lobule. Toxicol Appl Pharmacol 2023; 471:116550. [PMID: 37172768 PMCID: PMC10330769 DOI: 10.1016/j.taap.2023.116550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
The persistent environmental aryl hydrocarbon receptor agonist and hepatotoxin TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) induces hepatic lipid accumulation (steatosis), inflammation (steatohepatitis) and fibrosis. Thousands of liver-expressed, nuclear-localized lncRNAs with regulatory potential have been identified; however, their roles in TCDD-induced hepatoxicity and liver disease are unknown. We analyzed single nucleus (sn)RNA-seq data from control and subchronic (4 wk) TCDD-exposed mouse liver to determine liver cell-type specificity, zonation and differential expression profiles for thousands of lncRNAs. TCDD dysregulated >4000 of these lncRNAs in one or more liver cell types, including 684 lncRNAs specifically dysregulated in liver non-parenchymal cells. Trajectory inference analysis revealed major disruption by TCDD of hepatocyte zonation, affecting >800 genes, including 121 lncRNAs, with strong enrichment for lipid metabolism genes. TCDD also dysregulated expression of >200 transcription factors, including 19 Nuclear Receptors, most notably in hepatocytes and Kupffer cells. TCDD-induced changes in cell-cell communication patterns included marked decreases in EGF signaling from hepatocytes to non-parenchymal cells and increases in extracellular matrix-receptor interactions central to liver fibrosis. Gene regulatory networks constructed from the snRNA-seq data identified TCDD-exposed liver network-essential lncRNA regulators linked to functions such as fatty acid metabolic process, peroxisome and xenobiotic metabolism. Networks were validated by the striking enrichments that predicted regulatory lncRNAs showed for specific biological pathways. These findings highlight the power of snRNA-seq to discover functional roles for many xenobiotic-responsive lncRNAs in both hepatocytes and liver non-parenchymal cells and to elucidate novel aspects of foreign chemical-induced hepatotoxicity and liver disease, including dysregulation of intercellular communication within the liver lobule.
Collapse
Affiliation(s)
- Kritika Karri
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
10
|
Ding Y, Xu X, Meng B, Wang L, Zhu B, Guo B, Zhang J, Xiang L, Dong J, Liu M, Xiang G. Myeloid-derived growth factor alleviates non-alcoholic fatty liver disease alleviates in a manner involving IKKβ/NF-κB signaling. Cell Death Dis 2023; 14:376. [PMID: 37365185 DOI: 10.1038/s41419-023-05904-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/31/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023]
Abstract
Whether bone marrow modulates systemic metabolism remains unknown. Our recent study suggested that myeloid-derived growth factor (MYDGF) improves insulin resistance. Here, we found that myeloid cell-specific MYDGF deficiency aggravated hepatic inflammation, lipogenesis, and steatosis, and show that myeloid cell-derived MYDGF restoration alleviated hepatic inflammation, lipogenesis, and steatosis. Additionally, recombinant MYDGF attenuated inflammation, lipogenesis, and fat deposition in primary mouse hepatocytes (PMHs). Importantly, inhibitor kappa B kinase beta/nuclear factor-kappa B (IKKβ/NF-κB) signaling is involved in protection of MYDGF on non-alcoholic fatty liver disease (NAFLD). These data revealed that myeloid cell-derived MYDGF alleviates NAFLD and inflammation in a manner involving IKKβ/NF-κB signaling, and serves as a factor involved in the crosstalk between the liver and bone marrow that regulates liver fat metabolism. Bone marrow functions as an endocrine organ and serves as a potential therapeutic target for metabolic disorders.
Collapse
Affiliation(s)
- Yan Ding
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
- Department of Diagnostics, School of Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan Province, China
| | - Xiaoli Xu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, NO.1023, South Shatai Road, Guangzhou, 510515, Guangdong Province, China
| | - Biying Meng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, NO.1023, South Shatai Road, Guangzhou, 510515, Guangdong Province, China
| | - Li Wang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
| | - Biao Zhu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
| | - Bei Guo
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
| | - Jiajia Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
| | - Lin Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
| | - Jing Dong
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
| | - Min Liu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China
| | - Guangda Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, 430070, Hubei Province, China.
- The First School of Clinical Medicine, Southern Medical University, NO.1023, South Shatai Road, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|
11
|
Karri K, Waxman DJ. TCDD dysregulation of lncRNA expression, liver zonation and intercellular communication across the liver lobule. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.523119. [PMID: 36711947 PMCID: PMC9881922 DOI: 10.1101/2023.01.07.523119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The persistent environmental aryl hydrocarbon receptor agonist and hepatotoxin TCDD (2,3,7,8-tetrachlorodibenzo- p -dioxin) induces hepatic lipid accumulation (steatosis), inflammation (steatohepatitis) and fibrosis. Thousands of liver-expressed, nuclear-localized lncRNAs with regulatory potential have been identified; however, their roles in TCDD-induced hepatoxicity and liver disease are unknown. We analyzed single nucleus (sn)RNA-seq data from control and chronic TCDD-exposed mouse liver to determine liver cell-type specificity, zonation and differential expression profiles for thousands of IncRNAs. TCDD dysregulated >4,000 of these lncRNAs in one or more liver cell types, including 684 lncRNAs specifically dysregulated in liver non-parenchymal cells. Trajectory inference analysis revealed major disruption by TCDD of hepatocyte zonation, affecting >800 genes, including 121 IncRNAs, with strong enrichment for lipid metabolism genes. TCDD also dysregulated expression of >200 transcription factors, including 19 Nuclear Receptors, most notably in hepatocytes and Kupffer cells. TCDD-induced changes in cellâ€"cell communication patterns included marked decreases in EGF signaling from hepatocytes to non-parenchymal cells and increases in extracellular matrix-receptor interactions central to liver fibrosis. Gene regulatory networks constructed from the snRNA-seq data identified TCDD-exposed liver network-essential lncRNA regulators linked to functions such as fatty acid metabolic process, peroxisome and xenobiotic metabolic. Networks were validated by the striking enrichments that predicted regulatory IncRNAs showed for specific biological pathways. These findings highlight the power of snRNA-seq to discover functional roles for many xenobiotic-responsive lncRNAs in both hepatocytes and liver non-parenchymal cells and to elucidate novel aspects of foreign chemical-induced hepatotoxicity and liver disease, including dysregulation of intercellular communication within the liver lobule.
Collapse
|
12
|
Yoon JY, Kim JY, Kim HJ, Ka NL, Lee SH, Lee MO. LncRNA Ctcflos modulates glucocorticoid receptor-mediated induction of hepatic phosphoenolpyruvate carboxykinase in mice. Life Sci 2022; 312:121254. [PMID: 36470542 DOI: 10.1016/j.lfs.2022.121254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/16/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Jae-Yeun Yoon
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ju-Yeon Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Na-Lee Ka
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Heon Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea; Bio-MAX institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Tsermpini EE, Glamočlija U, Ulucan-Karnak F, Redenšek Trampuž S, Dolžan V. Molecular Mechanisms Related to Responses to Oxidative Stress and Antioxidative Therapies in COVID-19: A Systematic Review. Antioxidants (Basel) 2022; 11:1609. [PMID: 36009328 PMCID: PMC9405444 DOI: 10.3390/antiox11081609] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/25/2022] Open
Abstract
The coronavirus disease (COVID-19) pandemic is a leading global health and economic challenge. What defines the disease's progression is not entirely understood, but there are strong indications that oxidative stress and the defense against reactive oxygen species are crucial players. A big influx of immune cells to the site of infection is marked by the increase in reactive oxygen and nitrogen species. Our article aims to highlight the critical role of oxidative stress in the emergence and severity of COVID-19 and, more importantly, to shed light on the underlying molecular and genetic mechanisms. We have reviewed the available literature and clinical trials to extract the relevant genetic variants within the oxidative stress pathway associated with COVID-19 and the anti-oxidative therapies currently evaluated in the clinical trials for COVID-19 treatment, in particular clinical trials on glutathione and N-acetylcysteine.
Collapse
Affiliation(s)
- Evangelia Eirini Tsermpini
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Una Glamočlija
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina
- School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Fulden Ulucan-Karnak
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, 35100 İzmir, Turkey
| | - Sara Redenšek Trampuž
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
14
|
Han YH, Choi H, Kim HJ, Lee MO. Chemotactic cytokines secreted from Kupffer cells contribute to the sex-dependent susceptibility to non-alcoholic fatty liver diseases in mice. Life Sci 2022; 306:120846. [PMID: 35914587 DOI: 10.1016/j.lfs.2022.120846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022]
Abstract
AIMS The global prevalence of non-alcoholic fatty liver disease (NAFLD) has rapidly increased over the last decade due to an elevated occurrence of metabolic syndromes. Importantly, the prevalence and severity of NAFLD is higher in men than in women. Therefore, in the present study we endeavored to identify the mechanistic disparity between male and female mice. MAIN METHODS Global gene transcriptomics analysis was done with the high-fat diet (HFD)-induced NAFLD model of male, female, and ovariectomized (OVX) female mice. The expression of CCL2, CXCL2, and CXCL10 in mRNA level and serum protein level was done by qPCR and ELISA each. Immunohistochemistry staining was used to observe hepatic immune cell infiltration. To analyzing portion of immune cells, flow cytometry was done with isolated liver cells from HFD-fed male and female mice. Primary mouse liver cells were isolated from male and female mice for in vitro studies. KEY FINDINGS We identified sex differences in inflammatory chemokines, CCL2, CXCL2, and CXCL10, with the expression of these chemokines enhanced in male and OVX, but not in female, mice after HFD feeding. Resident Kupffer cells (KCs) were identified as the major source of production of CCL2, CXCL2, and CXCL10 in the mouse NAFLD model. Notably, KCs obtained from male mice expressed higher levels of chemokines than those from female mice, indicating that KCs may mediate the sex discrepancy in NAFLD progression. SIGNIFICANCE Our findings offer new insights into the pathology of sex-specific differences in NAFLD, involving chemokines and KCs.
Collapse
Affiliation(s)
- Yong-Hyun Han
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea.
| | - Haena Choi
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea; Bio-MAX institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Gu H, Hu P, Zhao Y, Liu Y, Wang YT, Ahmed AA, Liu HY, Cai D. Nuclear Receptor RORα/γ: Exciting Modulators in Metabolic Syndrome and Related Disorders. Front Nutr 2022; 9:925267. [PMID: 35799591 PMCID: PMC9253614 DOI: 10.3389/fnut.2022.925267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Abstract
Under the influences of modern lifestyle, metabolic syndromes (MetS), including insulin resistance, obesity, and fatty liver, featuring a worldwide chronic disease, greatly raise the risk of type 2 diabetes, heart disease, and stroke. However, its pathogenesis is still unclear, and there are limited drugs with strong clinical efficacy and specificity. Given the close connection between impaired lipid metabolism and MetS onset, modulating the lipid metabolic genes may provide potential prospects in the development of MetS therapeutics. Nuclear receptors are such druggable transcription factors that translate physiological signals into gene regulation via DNA binding upon ligand activation. Recent studies reveal vital functions of the NRs retinoic acid's receptor-related orphan receptors (RORs), including RORα and RORγ, in the gene regulation in lipid metabolism and MetS. This review focuses on the latest developments in their actions on MetS and related metabolic disorders, which would benefit future clinically therapeutic applications.
Collapse
Affiliation(s)
- Haotian Gu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yahui Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yaya Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yi-Ting Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Abdelkareem A. Ahmed
- Department of Veterinary Biomedical Sciences, Botswana University of Agriculture and Agriculture and Natural Resources, Gaborone, Botswana
- Biomedical Research Institute, Darfur University College, Nyala, Sudan
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- *Correspondence: Hao-Yu Liu
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Demin Cai
| |
Collapse
|
16
|
Song J, Song H, Wei H, Sun R, Tian Z, Peng H. Requirement of RORα for maintenance and antitumor immunity of liver-resident natural killer cells/ILC1s. Hepatology 2022; 75:1181-1193. [PMID: 34510508 DOI: 10.1002/hep.32147] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 08/24/2021] [Accepted: 09/07/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUD AND AIMS Liver type 1 innate lymphoid cells (ILC1s), also known as liver-resident natural killer (LrNK) cells, comprise a high proportion of total hepatic ILCs. However, factors regulating their maintenance and function remain unclear. APPROACH AND RESULTS In this study, we found high expression of retinoid-related orphan nuclear receptor alpha (RORα) in LrNK cells/ILC1s. Mice with conditional ablation of retinoid-related orphan nuclear receptor alpha (Rorα) in LrNK cells/ILC1s and conventional natural killer (cNK) cells had decreased LrNK cells/ILC1s but normal numbers of cNK cells. RORα-deficient LrNK cells/ILC1s displayed increased apoptosis and significantly altered transcriptional profile. Using a murine model of colorectal cancer liver metastasis, we found that RORα conditional deficiency resulted in more aggressive liver tumor progression and impaired effector molecule expression in LrNK cells/ILC1s. Consequently, treatment with the RORα agonist efficiently limited liver metastases and promoted effector molecule expression of LrNK cells/ILC1s. CONCLUSIONS This study reveals a role of RORα in LrNK cell/ILC1 maintenance and function, providing insights into the harnessing of LrNK cell/ILC1 activity in the treatment of liver cancer.
Collapse
Affiliation(s)
- Jiaxi Song
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina.,Institute of ImmunologyUniversity of Science and Technology of ChinaHefeiChina
| | - Hao Song
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina.,Institute of ImmunologyUniversity of Science and Technology of ChinaHefeiChina
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina.,Institute of ImmunologyUniversity of Science and Technology of ChinaHefeiChina
| | - Rui Sun
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina.,Institute of ImmunologyUniversity of Science and Technology of ChinaHefeiChina
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina.,Institute of ImmunologyUniversity of Science and Technology of ChinaHefeiChina.,Research Unit of NK Cell StudyChinese Academy of Medical SciencesHefeiChina
| | - Hui Peng
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina.,Institute of ImmunologyUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
17
|
Zhang X, Deng D, Cui D, Liu Y, He S, Zhang H, Xie Y, Yu X, Yang S, Chen Y, Su Z. Cholesterol Sulfate Exerts Protective Effect on Pancreatic β-Cells by Regulating β-Cell Mass and Insulin Secretion. Front Pharmacol 2022; 13:840406. [PMID: 35308228 PMCID: PMC8930834 DOI: 10.3389/fphar.2022.840406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/04/2022] [Indexed: 12/04/2022] Open
Abstract
Rational: Cholesterol sulfate (CS) is the most abundant known sterol sulfate in human plasma, and it plays a significant role in the control of metabolism and inflammatory response, which contribute to the pathogenesis of insulin resistance, β-cell dysfunction and the resultant development of diabetes. However, the role of CS in β-cells and its effect on the development of diabetes remain unknown. Here, we determined the physiological function of CS in pancreatic β-cell homeostasis. Materials and Methods: Blood CS levels in streptozotocin (STZ)- or high-fat diet-induced diabetic mice and patients with type 1 or 2 diabetes were determined by LC-MS/MS. The impact of CS on β-cell mass and insulin secretion was investigated in vitro in isolated mouse islets and the β-cell line INS-1 and in vivo in STZ-induced diabetic mice. The molecular mechanism of CS was explored by viability assay, EdU incorporation analysis, flow cytometry, intracellular Ca2+ influx analysis, mitochondrial membrane potential and cellular ROS assays, and metabolism assay kits. Results: Plasma CS levels in mice and humans were significantly elevated under diabetic conditions. CS attenuated diabetes in a low-dose STZ-induced mouse model. Mechanistically, CS promoted β-cell proliferation and protected β-cells against apoptosis under stressful conditions, which in turn preserved β-cell mass. In addition, CS supported glucose transporter-2 (GLUT2) expression and mitochondrial integrity, which then resulted in a less reactive oxygen species (ROS) generation and an increase in ATP production, thereby enabling insulin secretion machinery in the islets to function adequately. Conclusion: This study revealed a novel dual role of CS in integrating β-cell survival and cell function, suggesting that CS might offer a physiologic approach to preserve β-cells and protect against the development of diabetes mellitus.
Collapse
Affiliation(s)
- Xueping Zhang
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Dan Deng
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Daxin Cui
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yin Liu
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Siyuan He
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Hongmei Zhang
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yaorui Xie
- Department of Clinical Laboratory, Sichuan Provincial Peoples Hospital Jinniu Hospital, Chengdu, China
| | - Xiaoqian Yu
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Shanshan Yang
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yulong Chen
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Zhiguang Su
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
- *Correspondence: Zhiguang Su,
| |
Collapse
|
18
|
Xiao L, Wang M, Zhang W, Song Y, Zeng J, Li H, Yu H, Li L, Gao P, Yao P. Maternal diabetes-mediated RORA suppression contributes to gastrointestinal symptoms in autism-like mouse offspring. BMC Neurosci 2022; 23:8. [PMID: 35164690 PMCID: PMC8842926 DOI: 10.1186/s12868-022-00693-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Retinoic acid-related orphan receptor alpha (RORA) has been reported to be suppressed in autistic patients and is associated with autism spectrum disorders (ASD), although the potential role and mechanism of RORA on gastrointestinal (GI) symptoms in ASD patients is still not reported. In this study, we aim to investigate the contribution of RORA to GI symptoms through a maternal diabetes-mediated autism-like mouse model. RESULTS Male offspring of diabetic dams were treated with either superoxide dismutase (SOD) mimetic MnTBAP or RORA agonist SR1078, or were crossbred with intestine epithelial cells (IEC)-specific RORA knockout (RORA-/-) mouse. Gene expression, oxidative stress and inflammation were measured in brain tissues, peripheral blood mononuclear cells (PBMC) and IEC, and GI symptoms were evaluated. Our results showed that SOD mimetic MnTBAP completely, while RORA agonist SR1078 partly, reversed maternal diabetes-mediated oxidative stress and inflammation in the brain, PBMC and IEC, as well as GI symptoms, including intestine permeability and altered gut microbiota compositions. IEC-specific RORA deficiency either mimicked or worsened maternal diabetes-mediated GI symptoms as well as oxidative stress and inflammation in IEC, while there was little effect on maternal diabetes-mediated autism-like behaviors. CONCLUSIONS We conclude that RORA suppression contributes to maternal diabetes-mediated GI symptoms in autism-like mouse offspring, this study provides a potential therapeutical target for maternal diabetes-mediated GI symptoms in offspring through RORA activation.
Collapse
Affiliation(s)
- Li Xiao
- Department of Pediatrics, Affiliated Foshan Maternity & Child Healthcare Hospital, The Second School of Clinical Medicine of Southern Medical University, Foshan, 528000, People's Republic of China
| | - Min Wang
- Hainan Women and Children's Medical Center, Haikou, 570206, People's Republic of China
| | - Wanhua Zhang
- Department of Pediatrics, Affiliated Foshan Maternity & Child Healthcare Hospital, The Second School of Clinical Medicine of Southern Medical University, Foshan, 528000, People's Republic of China
| | - Yuan Song
- Hainan Women and Children's Medical Center, Haikou, 570206, People's Republic of China
| | - Jiaying Zeng
- Department of Pediatrics, Affiliated Foshan Maternity & Child Healthcare Hospital, The Second School of Clinical Medicine of Southern Medical University, Foshan, 528000, People's Republic of China
| | - Huilin Li
- Hainan Women and Children's Medical Center, Haikou, 570206, People's Republic of China
| | - Hong Yu
- Department of Pediatrics, Affiliated Foshan Maternity & Child Healthcare Hospital, The Second School of Clinical Medicine of Southern Medical University, Foshan, 528000, People's Republic of China
| | - Ling Li
- Hainan Women and Children's Medical Center, Haikou, 570206, People's Republic of China.
| | - Pingming Gao
- Department of Pediatrics, Affiliated Foshan Maternity & Child Healthcare Hospital, The Second School of Clinical Medicine of Southern Medical University, Foshan, 528000, People's Republic of China.
| | - Paul Yao
- Department of Pediatrics, Affiliated Foshan Maternity & Child Healthcare Hospital, The Second School of Clinical Medicine of Southern Medical University, Foshan, 528000, People's Republic of China. .,Hainan Women and Children's Medical Center, Haikou, 570206, People's Republic of China.
| |
Collapse
|
19
|
Kim JY, Yang IS, Kim HJ, Yoon JY, Han YH, Seong JK, Lee MO. RORα contributes to the maintenance of genome ploidy in the liver of mice with diet-induced nonalcoholic steatohepatitis. Am J Physiol Endocrinol Metab 2022; 322:E118-E131. [PMID: 34894722 DOI: 10.1152/ajpendo.00309.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hepatic polyploidization is closely linked to the progression of nonalcoholic fatty liver disease (NAFLD); however, the underlying molecular mechanism is not clearly understood. In this study, we demonstrated the role of retinoic acid-related orphan receptor α (RORα) in the maintenance of genomic integrity, particularly in the pathogenesis of NAFLD, using the high-fat diet (HFD)-fed liver-specific RORα knockout (RORα-LKO) mouse model. First, we observed that the loss of hepatic retinoic acid receptor-related orphan receptor α (RORα) accelerated hepatocyte nuclear polyploidization after HFD feeding. In 70% partial hepatectomy experiments, enrichment of hepatocyte polyploidy was more obvious in the RORα-LKO animals, which was accompanied by early progression to the S phase and blockade of the G2/M transition, suggesting a potential role of RORα in suppressing hepatocyte polyploidization in the regenerating liver. An analysis of a publicly available RNA sequencing (RNA-seq) and chromatin immunoprecipitation-seq dataset, together with the Search Tool of the Retrieval of Interacting Genes/Proteins database resource, revealed that DNA endoreplication was the top-enriched biological process Gene Ontology term. Furthermore, we found that E2f7 and E2f8, which encode key transcription factors for DNA endoreplication, were the downstream targets of RORα-induced transcriptional repression. Finally, we showed that the administration of JC1-40, an RORα activator (5 mg/kg body wt), significantly reduced hepatic nuclear polyploidization in the HFD-fed mice. Together, our observations suggest that the RORα-induced suppression of hepatic polyploidization may provide new insights into the pathological polyploidy of NAFLD and may contribute to the development of therapeutic strategies for the treatment of NAFLD.NEW & NOTEWORTHY It has been reported that hepatic polyploidization is closely linked to the progression of NAFLD. Here, we showed that the genetic depletion of hepatic RORα in mice accelerated hepatocyte polyploidization after high-fat diet feeding. The mechanism could be the RORα-mediated repression of E2f7 and E2f8, key transcription factors for DNA endoreplication. Thus, preservation of genome integrity by RORα could provide a new insight for developing therapeutics against the disease.
Collapse
Affiliation(s)
- Ju-Yeon Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - In Sook Yang
- College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Jae-Yeun Yoon
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Yong-Hyun Han
- College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Je Kyung Seong
- College of Veterinary Medicine, Seoul National University, Seoul, South Korea
- Research Institute of Veterinary Science, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, South Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
- Bio-MAX Institute, Seoul National University, Seoul, South Korea
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, South Korea
| |
Collapse
|
20
|
Kim H, Han Y, Kim J, Lee M. RORα Enhances Lysosomal Acidification and Autophagic Flux in the Hepatocytes. Hepatol Commun 2021; 5:2121-2138. [PMID: 34558854 PMCID: PMC8631090 DOI: 10.1002/hep4.1785] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/25/2021] [Accepted: 06/24/2021] [Indexed: 12/30/2022] Open
Abstract
Lysosomes are intracellular acidic organelles with catabolic functions that contribute to the activation of autophagy. Although autophagy abnormality is associated with defects in lysosomal acidification during the progression of nonalcoholic fatty liver disease (NAFLD), the mechanisms of control of lysosomal acidification are not well understood at the molecular level. Thus, we aimed to elucidate the role of the orphan nuclear receptor retinoic acid-related orphan receptor α (RORα) in lysosomal acidification and autophagic flux, particularly in nutrition-enriched hepatocytes. First, lysosomal acidity was much lower in the hepatocytes obtained from hepatocyte-specific RORα-deleted (RORα-LKO) mice, whereas the infusion of an adenovirus encoding RORα in wild-type hepatocytes increased lysosomal acidity, as determined by LysoSensor. Second, the lysosomal translocation of the mechanistic target of rapamycin was increased and immature cathepsin D was accumulated in the liver of RORα-LKO mice. Third, the accumulation of LC3-II, p62/sequestosome 1 (SQSTM1), and neighbor of BRCA1 gene 1 (NBR1) was increased in the livers of RORα-LKO mice, indicating an impaired autophagic flux in the livers. Consistently, the number of autolysosomes containing mitochondria and lipid droplets was dramatically reduced in the RORα-deleted hepatocytes. Finally, we found that RORα induced the transcription of genes involved in lysosomal function, such as Atp6v1g1, a vacuolar H+ -ATPase (v-ATPase) subunit, which were largely down-regulated in the livers of mice with high-fat diet-induced NAFLD and patients with hepatitis. Conclusion: Targeting RORα may be a potential therapeutic strategy to restore lysosomal acidification, which inhibits the progression of NAFLD.
Collapse
Affiliation(s)
- Hyeon‐Ji Kim
- College of PharmacySeoul National UniversitySeoulKorea
| | - Yong‐Hyun Han
- Laboratory of Pathology and PhysiologyCollege of PharmacyKangwon National UniversityChuncheonSouth Korea
| | - Ju‐Yeon Kim
- College of PharmacySeoul National UniversitySeoulKorea
| | - Mi‐Ock Lee
- College of PharmacySeoul National UniversitySeoulKorea
- Bio‐MAX InstituteSeoul National UniversitySeoulKorea
- Research Institute of Pharmaceutical SciencesSeoulKorea
| |
Collapse
|
21
|
TM4SF5-dependent crosstalk between hepatocytes and macrophages to reprogram the inflammatory environment. Cell Rep 2021; 37:110018. [PMID: 34788612 DOI: 10.1016/j.celrep.2021.110018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/10/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic injury to hepatocytes results in inflammation, steatohepatitis, fibrosis, and nonalcoholic fatty liver disease (NAFLD). The tetraspanin TM4SF5 is implicated in fibrosis and cancer. We investigate the role of TM4SF5 in communication between hepatocytes and macrophages (MΦs) and its possible influence on the inflammatory microenvironment that may lead to NAFLD. TM4SF5 induction in differentiated MΦs promotes glucose uptake, glycolysis, and glucose sensitivity, leading to M1-type MΦ activation. Activated M1-type MΦs secrete pro-inflammatory interleukin-6 (IL-6), which induces the secretion of CCL20 and CXCL10 from TM4SF5-positive hepatocytes. Although TM4SF5-dependent secretion of these chemokines enhances glycolysis in M0 MΦs, further chronic exposure reprograms MΦs for an increase in the proportion of M2-type MΦs in the population, which may support diet- and chemical-induced NAFLD progression. We suggest that TM4SF5 expression in MΦs and hepatocytes is critically involved in modulating the inflammatory environment during NAFLD progression.
Collapse
|
22
|
The Role and Mechanism of Oxidative Stress and Nuclear Receptors in the Development of NAFLD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6889533. [PMID: 34745420 PMCID: PMC8566046 DOI: 10.1155/2021/6889533] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
The overproduction of reactive oxygen species (ROS) and consequent oxidative stress contribute to the pathogenesis of acute and chronic liver diseases. It is now acknowledged that nonalcoholic fatty liver disease (NAFLD) is characterized as a redox-centered disease due to the role of ROS in hepatic metabolism. However, the underlying mechanisms accounting for these alternations are not completely understood. Several nuclear receptors (NRs) are dysregulated in NAFLD, and have a direct influence on the expression of a set of genes relating to the progress of hepatic lipid homeostasis and ROS generation. Meanwhile, the NRs act as redox sensors in response to metabolic stress. Therefore, targeting NRs may represent a promising strategy for improving oxidation damage and treating NAFLD. This review summarizes the link between impaired lipid metabolism and oxidative stress and highlights some NRs involved in regulating oxidant/antioxidant turnover in the context of NAFLD, shedding light on potential therapies based on NR-mediated modulation of ROS generation and lipid accumulation.
Collapse
|
23
|
Matsuoka H, Michihara A. Identification of the RORα Transcriptional Network Contributes to the Search for Therapeutic Targets in Atherosclerosis. Biol Pharm Bull 2021; 44:1607-1616. [PMID: 34719639 DOI: 10.1248/bpb.b21-00426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The retinoic acid receptor-related orphan receptor α (RORα) is involved in the regulation of several physiological processes, including development, metabolism, and circadian rhythm. RORα-deficient mice display profound atherosclerosis, in which hypoalphalipoproteinemia is reportedly associated with decreased plasma levels of high-density lipoprotein, increased levels of inflammatory cytokines, and ischemia/reperfusion-induced damage. The recent characterization of endogenous ligands (including cholesterol, oxysterols, provitamin D3, and their derivatives), mediators, and initiation complexes associated with the transcriptional regulation of these orphan nuclear receptors has facilitated the development of synthetic ligands. These findings have also highlighted the potential of application of RORα as a therapeutic target for several diseases, including diabetes, dyslipidemia, and atherosclerosis. In this review, the current literature related to the structure and function of RORα, its genetic inter-individual differences, and its potential as a therapeutic target in atherosclerosis is discussed.
Collapse
Affiliation(s)
- Hiroshi Matsuoka
- Laboratory of Genomic Function and Pathophysiology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University
| | - Akihiro Michihara
- Laboratory of Genomic Function and Pathophysiology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University
| |
Collapse
|
24
|
Sen A, Anakk S. Jekyll and Hyde: nuclear receptors ignite and extinguish hepatic oxidative milieu. Trends Endocrinol Metab 2021; 32:790-802. [PMID: 34481730 PMCID: PMC8464172 DOI: 10.1016/j.tem.2021.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/21/2022]
Abstract
Nuclear receptors (NRs) are ligand-binding transcription factors that regulate gene networks and physiological responses. Often oxidative stress precedes the onset of liver diseases, and Nrf2 is a key regulator of antioxidant pathways. NRs crosstalk with Nrf2, since NR activation can influence the oxidative milieu by modulating reductive cellular processes. Diet and xenobiotics also regulate NR expression and activity, suggesting a feedback loop. Depending on the tissue context and cues, NRs either increase or decrease toxicity and oxidative damage. Many FDA-approved drugs target NRs, and one could potentially repurpose them to ameliorate reactive oxygen species (ROS). Here, we discuss how several NRs modulate oxidative stress subsequent to diet, organic pollutants, and drug-induced injury to the liver.
Collapse
Affiliation(s)
- Anushna Sen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
25
|
Mao W, Xiong G, Wu Y, Wang C, St. Clair D, Li JD, Xu R. RORα Suppresses Cancer-Associated Inflammation by Repressing Respiratory Complex I-Dependent ROS Generation. Int J Mol Sci 2021; 22:ijms221910665. [PMID: 34639006 PMCID: PMC8509002 DOI: 10.3390/ijms221910665] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Breast cancer development is associated with macrophage infiltration and differentiation in the tumor microenvironment. Our previous study highlights the crucial function of reactive oxygen species (ROS) in enhancing macrophage infiltration during the disruption of mammary tissue polarity. However, the regulation of ROS and ROS-associated macrophage infiltration in breast cancer has not been fully determined. Previous studies identified retinoid orphan nuclear receptor alpha (RORα) as a potential tumor suppressor in human breast cancer. In the present study, we showed that retinoid orphan nuclear receptor alpha (RORα) significantly decreased ROS levels and inhibited ROS-mediated cytokine expression in breast cancer cells. RORα expression in mammary epithelial cells inhibited macrophage infiltration by repressing ROS generation in the co-culture assay. Using gene co-expression and chromatin immunoprecipitation (ChIP) analyses, we identified complex I subunits NDUFS6 and NDUFA11 as RORα targets that mediated its function in suppressing superoxide generation in mitochondria. Notably, the expression of RORα in 4T1 cells significantly inhibited cancer metastasis, reduced macrophage accumulation, and enhanced M1-like macrophage differentiation in tumor tissue. In addition, reduced RORα expression in breast cancer tissue was associated with an increased incidence of cancer metastasis. These results provide additional insights into cancer-associated inflammation, and identify RORα as a potential target to suppress ROS-induced mammary tumor progression.
Collapse
Affiliation(s)
- Wei Mao
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha 410078, China;
| | - Gaofeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Yuanyuan Wu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Daret St. Clair
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jia-Da Li
- Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha 410078, China;
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (W.M.); (G.X.); (Y.W.); (C.W.); (D.S.C.)
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-859-323-7889
| |
Collapse
|
26
|
Xiang K, Xu Z, Hu YQ, He YS, Wu GC, Li TY, Wang XR, Ding LH, Zhang Q, Tao SS, Ye DQ, Pan HF, Wang DG. Circadian clock genes as promising therapeutic targets for autoimmune diseases. Autoimmun Rev 2021; 20:102866. [PMID: 34118460 DOI: 10.1016/j.autrev.2021.102866] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 03/30/2021] [Indexed: 12/31/2022]
Abstract
Circadian rhythm is a natural, endogenous process whose physiological functions are controlled by a set of clock genes. Disturbance of the clock genes have detrimental effects on both innate and adaptive immunity, which significantly enhance pro-inflammatory responses and susceptibility to autoimmune diseases via strictly controlling the individual cellular components of the immune system that initiate and perpetuate the inflammation pathways. Autoimmune diseases, especially rheumatoid arthritis (RA), often exhibit substantial circadian oscillations, and circadian rhythm is involved in the onset and progression of autoimmune diseases. Mounting evidence indicate that the synthetic ligands of circadian clock genes have the property of reducing the susceptibility and clinical severity of subjects. This review supplies an overview of the roles of circadian clock genes in the pathology of autoimmune diseases, including BMAL1, CLOCK, PER, CRY, REV-ERBα, and ROR. Furthermore, summarized some circadian clock genes as candidate genes for autoimmune diseases and current advancement on therapy of autoimmune diseases with synthetic ligands of circadian clock genes. The existing body of knowledge demonstrates that circadian clock genes are inextricably linked to autoimmune diseases. Future research should pay attention to improve the quality of life of patients with autoimmune diseases and reduce the effects of drug preparation on the normal circadian rhythms.
Collapse
Affiliation(s)
- Kun Xiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Zhiwei Xu
- School of Public Health, Faculty of Medicine, University of Queensland, 288 Herston Road, Herston, QLD, 4006, Brisbane, Australia
| | - Yu-Qian Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Guo-Cui Wu
- School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Tian-Yu Li
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xue-Rong Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Li-Hong Ding
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qin Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Sha-Sha Tao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China.
| | - De-Guang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
27
|
Wang SW, Lan T, Sheng H, Zheng F, Lei MK, Wang LX, Chen HF, Xu CY, Zhang F. Nobiletin Alleviates Non-alcoholic Steatohepatitis in MCD-Induced Mice by Regulating Macrophage Polarization. Front Physiol 2021; 12:687744. [PMID: 34093242 PMCID: PMC8174844 DOI: 10.3389/fphys.2021.687744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is an inflammatory disorder that is characterized by chronic activation of the hepatic inflammatory response and subsequent liver damage. The regulation of macrophage polarization in liver is closely related to the progression of NASH. The orphan nuclear receptor retinoic-acid-related orphan receptor α (RORα) and Krüppel-like factor 4 (KLF4) are key regulators which promote hepatic macrophages toward M2 phenotype and protect against NASH in mice. Nobiletin (NOB), a natural polymethoxylated flavone, is previously reported as a RORα regulator in diet-induced obese mice. However, it is still unclear whether NOB has the protective effect on NASH. In this study, we investigated the role of NOB in NASH using a methionine and choline deficient (MCD)-induced NASH mouse model. Our results showed that NOB ameliorated hepatic damage and fibrosis in MCD fed mice. NOB treatment reduced the infiltration of macrophages and neutrophils in the liver in MCD-fed mice. Of importance, NOB significantly increased the proportion of M2 macrophages and the expression of anti-inflammatory factors in vivo and in vitro. Meanwhile, NOB also decreased the population of M1 macrophages and the expression of proinflammatory cytokines. Mechanistically, NOB elevated KLF4 expression in macrophages. Inhibition of KLF4 abolished NOB regulated macrophage polarization. Furthermore, the regulation of NOB in KLF4 expression was dependent on RORα.
Collapse
Affiliation(s)
- Si-Wei Wang
- Core Facility, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Tian Lan
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Hao Sheng
- Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Zheng
- Core Facility, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Mei-Kang Lei
- Department of Analytical Testing Center, Quzhou Customs, Quzhou, China
| | - Li-Xia Wang
- Agriculture and Rural Bureau of Changshan County, Quzhou, China
| | - Hang-Fei Chen
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Chun-Yi Xu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Feng Zhang
- Core Facility, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.,Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
28
|
Guillemot-Legris O, Muccioli GG. The oxysterome and its receptors as pharmacological targets in inflammatory diseases. Br J Pharmacol 2021; 179:4917-4940. [PMID: 33817775 DOI: 10.1111/bph.15479] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Oxysterols have gained attention over the last decades and are now considered as fully fledged bioactive lipids. The study of their levels in several conditions, including atherosclerosis, obesity and neurodegenerative diseases, led to a better understanding of their involvement in (patho)physiological processes such as inflammation and immunity. For instance, the characterization of the cholesterol-7α,25-dihydroxycholesterol/GPR183 axis and its implication in immunity represents an important step in the oxysterome study. Besides this axis, others were identified as important in several inflammatory pathologies (such as colitis, lung inflammation and atherosclerosis). However, the oxysterome is a complex system notably due to a redundancy of metabolic enzymes and a wide range of receptors. Indeed, deciphering oxysterol roles and identifying the potential receptor(s) involved in a given pathology remain challenging. Oxysterol properties are very diverse, but most of them could be connected by a common component: inflammation. Here, we review the implication of oxysterol receptors in inflammatory diseases.
Collapse
Affiliation(s)
- Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
29
|
Navigatore Fonzo L, Alfaro M, Mazaferro P, Golini R, Jorge L, Cecilia Della Vedova M, Ramirez D, Delsouc B, Casais M, Anzulovich AC. An intracerebroventricular injection of amyloid-beta peptide (1-42) aggregates modifies daily temporal organization of clock factors expression, protein carbonyls and antioxidant enzymes in the rat hippocampus. Brain Res 2021; 1767:147449. [PMID: 33771518 DOI: 10.1016/j.brainres.2021.147449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/26/2021] [Accepted: 03/19/2021] [Indexed: 10/21/2022]
Abstract
Alzheimer disease (AD) is the most frequent form of dementia in the elderly. It is characterized by the deterioration of memory and learning. The histopathological hallmarks of AD include the presence of extracellular deposits of amyloid beta peptide, intracellular neurofibrillary tangles, neuron and synapse loss, in the brain, including the hippocampus. Accumulation of Aβ peptide causes an increase in intracellular reactive oxygen species (ROS) and free radicals associated to a deficient antioxidant defense system. Besides oxidative stress and cognitive deficit, AD patients show alterations in their circadian rhythms. The objective of this work was to investigate the effects of an intracerebroventricular injection of amyloid beta peptide Aβ(1-42) aggregates on temporal patterns of protein oxidation, antioxidant enzymes and clock factors in the rat hippocampus. Four-month-old male Holtzman rats divided into the groups control (CO) and Aβ-injected (Aβ), were maintained under 12 h-light12h-dark conditions and received water and food ad-libitum. Hippocampus samples were isolated every 6 h during a 24 h period. Our results showed daily patterns of protein carbonyls, catalase (CAT) and glutathione peroxidase (GPx) expression and activity, as well as Rorα and Rev-erbß mRNA, in the rat hippocampus. Interestingly, an intracerebroventricular injection of Aβ aggregates modified daily oscillation of protein carbonyls levels, phase-shifted daily rhythms of clock genes and had a differential effect on the daily expression and activity of CAT and GPx. Thus, Aβ aggregates might affect clock-mediated transcriptional regulation of antioxidant enzymes, by affecting the formation of BMAL1:CLOCK heterodimer, probably, as a consequence of the alteration of the redox state observed in rats injected with Aβ.
Collapse
Affiliation(s)
| | - Mauro Alfaro
- Laboratory of Chronobiology, Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina
| | - Paula Mazaferro
- Laboratory of Chronobiology, Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina
| | - Rebeca Golini
- Laboratory of Chronobiology, Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina
| | - Leporatti Jorge
- Faculty of Economic, Legal and Social Sciences, National University of San Luis (UNSL), Campus Universitario, Ruta Prov. N° 55 (Ex. 148) Extremo Norte, D5700HHW San Luis, Argentina
| | - Maria Cecilia Della Vedova
- Institute of Chemistry-San Luis,(INQUISAL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina
| | - Darío Ramirez
- Laboratory of Experimental & Translational Medicine, Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina
| | - Belén Delsouc
- Laboratory of Biology Reproduction, Multidisciplinary Institute of Biological Res-earch-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina
| | - Marilina Casais
- Laboratory of Biology Reproduction, Multidisciplinary Institute of Biological Res-earch-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina
| | - Ana Cecilia Anzulovich
- Laboratory of Chronobiology, Av Ejército de los Andes N° 950, D5700HHW San Luis, Argentina.
| |
Collapse
|
30
|
Han YH, Kim HJ, Lee MO. RORα regulates hepatic lipolysis by inducing transcriptional expression of PNPLA3 in mice. Mol Cell Endocrinol 2021; 522:111122. [PMID: 33347955 DOI: 10.1016/j.mce.2020.111122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/16/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022]
Abstract
Nonalcoholic fatty liver diseases (NAFLDs) are characterized by excessive triacylglycerol (TAG) accumulation in the liver which contributes to hepatocyte dysfunction, inflammation, and fibrosis. Patatin-like phospholipase domain-containing 3 (PNPLA3; also known as adiponutrin) has emerged as an important enzyme leading to hepatic TAG hydrolysis. Because the I148M substitution in the PNPLA3 gene markedly reduces hepatic TAG hydrolase activity, this genetic variation is strongly associated with increased hepatic TAG in the full spectrum of NAFLDs. The Retinoic acid-related orphan receptor α (RORα) regulates various target genes related to lipid metabolism. Here, we investigated the role of RORα on PNPLA3-mediated hepatic lipid hydrolysis. With blockade of lipid esterification and β-oxidation, RORα enhanced TAG hydrolysis, resulting in increased free glycerol levels. We found a putative RORα response element on the upstream of PNPLA3 gene that was activated by RORα. Furthermore, the inhibitory action of cJUN on the RORα/PNPLA3 axis was enhanced under lipid stress and contributed to hepatic lipid accumulation. In summary, we showed for the first time that RORα activates the transcription of PNPLA3, which suggests that RORα and its ligands represent potential precision therapeutic approaches for NAFLDs.
Collapse
Affiliation(s)
- Yong-Hyun Han
- College of Pharmacy, Seoul National University, Seoul, South Korea; Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea; Bio-MAX Institute, Seoul National University, Seoul, South Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
31
|
|
32
|
L'homme L, Sermikli BP, Molendi-Coste O, Fleury S, Quemener S, Le Maître M, Joseph ML, Pineau L, Duhem C, Gross B, Vallez E, Tailleux A, Staels B, Dombrowicz D. Deletion of the nuclear receptor RORα in macrophages does not modify the development of obesity, insulin resistance and NASH. Sci Rep 2020; 10:21095. [PMID: 33273527 PMCID: PMC7713245 DOI: 10.1038/s41598-020-77858-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/02/2020] [Indexed: 11/09/2022] Open
Abstract
Retinoic acid receptor-related orphan receptor-alpha (RORα) is a transcription factor from the nuclear receptor family expressed by immune cells and involved in the development of obesity, insulin resistance (IR) and non-alcoholic steatohepatitis (NASH). It was recently reported that mice deficient for RORα in macrophages develop more severe NASH upon high fat diet (HFD) feeding due to altered Kupffer cell function. To better understand the role of RORα in obesity and IR, we independently generated a macrophage RORα-deficient mouse line. We report that RORα deletion in macrophages does not impact on HFD-induced obesity and IR. Surprisingly, we did not confirm an effect on NASH development upon HFD feeding nor in the more severe and obesity-independent choline-deficient, L-amino acid-defined diet model. Our results therefore show that RORα deletion in macrophages does not alter the development of obesity and IR and question its role in NASH.
Collapse
Affiliation(s)
- Laurent L'homme
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Benan Pelin Sermikli
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Olivier Molendi-Coste
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Sébastien Fleury
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Sandrine Quemener
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Mathilde Le Maître
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Marie-Laure Joseph
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Laurent Pineau
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Christian Duhem
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Barbara Gross
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Emmanuelle Vallez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Anne Tailleux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - David Dombrowicz
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France.
| |
Collapse
|
33
|
Cai X, Zhang P, Wang S, Hong L, Yu S, Li B, Zeng H, Yang X, Shao L. lncRNA FGD5 antisense RNA 1 upregulates RORA to suppress hypoxic injury of human cardiomyocyte cells by inhibiting oxidative stress and apoptosis via miR‑195. Mol Med Rep 2020; 22:4579-4588. [PMID: 33174051 PMCID: PMC7646841 DOI: 10.3892/mmr.2020.11558] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
Abstract
FGD5 antisense RNA 1 (FGD5-AS1) is a long non-coding RNA in acute myocardial infarction (AMI), which is primarily caused by myocardial ischemia-hypoxia. Retinoid acid receptor-related orphan receptor α (RORA) is a key protector in maintaining heart function. However, the roles of FGD5-AS1 and RORA in AMI have not previously been elucidated. The present study investigated the effect and mechanism of FGD5-AS1 and RORA in human cardiomyocyte AC16 cells under hypoxia. Reverse transcription-quantitative PCR and western blotting demonstrated that FGD5-AS1 and RORA were downregulated in the serum of patients with AMI and hypoxia-challenged AC16 cells. Functional experiments were performed via assays, flow cytometry and western blotting. In response to hypoxia, superoxide dismutase (SOD) activity was inhibited, but apoptosis rate and levels of reactive oxygen species and malondialdehyde were promoted in AC16 cells, accompanied by increased Bax and cleaved caspase-3 expression levels, and decreased SOD2 and glutathione peroxidase 1 expression levels. However, hypoxia-induced oxidative stress and apoptosis in AC16 cells were attenuated by ectopic expression of FGD5-AS1 or RORA. Moreover, silencing RORA counteracted the suppressive role of FGD5-AS1 overexpression in hypoxic injury. FGD5-AS1 controlled RORA expression levels via microRNA-195-5p (miR-195), as confirmed by dual-luciferase reporter and RNA pull-down assays. Consistently, miR-195 knockdown suppressed hypoxia-induced oxidative stress and apoptosis in AC16 cells, which was abrogated by downregulating FGD5-AS1 or RORA. In conclusion, FGD5-AS1 modulated hypoxic injury in human cardiomyocytes partially via the miR-195/RORA axis, suggesting FGD5-AS1 as a potential target in interfering with the progression of AMI.
Collapse
Affiliation(s)
- Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Zhang
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shu Wang
- Department of Gerontology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Songping Yu
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Li
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hong Zeng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xu Yang
- Shenzhen Realomics (Biotech), Co., Ltd., Shenzhen, Guangdong 518000, P.R. China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
34
|
Sanchez LD, Pontini L, Marinozzi M, Sanchez-Aranguren LC, Reis A, Dias IHK. Cholesterol and oxysterol sulfates: Pathophysiological roles and analytical challenges. Br J Pharmacol 2020; 178:3327-3341. [PMID: 32762060 DOI: 10.1111/bph.15227] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 01/18/2023] Open
Abstract
Cholesterol and oxysterol sulfates are important regulators of lipid metabolism, inflammation, cell apoptosis, and cell survival. Among the sulfate-based lipids, cholesterol sulfate (CS) is the most studied lipid both quantitatively and functionally. Despite the importance, very few studies have analysed and linked the actions of oxysterol sulfates to their physiological and pathophysiological roles. Overexpression of sulfotransferases confirmed the formation of a range of oxysterol sulfates and their antagonistic effects on liver X receptors (LXRs) prompting further investigations how are the changes to oxysterol/oxysterol sulfate homeostasis can contribute to LXR activity in the physiological milieu. Here, we aim to bring together for novel roles of oxysterol sulfates, the available techniques and the challenges associated with their analysis. Understanding the oxysterol/oxysterol sulfate levels and their pathophysiological mechanisms could lead to new therapeutic targets for metabolic diseases. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
| | - Lorenzo Pontini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maura Marinozzi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Ana Reis
- LAQV/REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | | |
Collapse
|
35
|
RORα autoregulates its transcription via MLL4-associated enhancer remodeling in the liver. Life Sci 2020; 256:118007. [DOI: 10.1016/j.lfs.2020.118007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/12/2020] [Accepted: 06/22/2020] [Indexed: 01/01/2023]
|
36
|
Zhang CH, Xiao Q, Sheng JQ, Liu TT, Cao YQ, Xue YN, Shi M, Cao Z, Zhou LF, Luo XQ, Deng KZ, Chen C. Gegen Qinlian Decoction abates nonalcoholic steatohepatitis associated liver injuries via anti-oxidative stress and anti-inflammatory response involved inhibition of toll-like receptor 4 signaling pathways. Biomed Pharmacother 2020; 126:110076. [PMID: 32169759 DOI: 10.1016/j.biopha.2020.110076] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023] Open
Abstract
Gegen Qilian Decoction (GGQLD) is a well-established classic Chinese medicine prescription in treating nonalcoholic steatohepatitis (NASH). However, the molecular mechanism of GGQLD action on NASH is still not clear. This study aimed to assess the anti-NASH effect of GGQLD, and to explore its molecular mechanisms in vivo and in vitro. In HFD-fed rats, GGQLD decreased significantly serum triglyceride (TG), cholesterol (CHO), total bile acid (TBA), low-density lipoprotein (LDL), free fatty acid (FFA) and lipopolysaccharide (LPS) levels, increased levels of differentially expressed proteins (DEPs) Ahcy, Gpx1, Mat1a, GNMT, and reduced the expression of ALDOB. In RAW264.7 macrophages, GGQLD reduced the expression levels of inflammatory factors TNF-α and IL-6 mRNA, and diminished NASH by increasing differentially expressed genes (DEGs) CBS, Mat1a, Hnf4α and Pparα to reduce oxidative stress or lipid metabolism. The results of DEGs verification also showed that GGQLD up-regulated expressions of Hnf4α, Pparα and Cbs genes. In HepG2 cells, GGQLD decreased IL-6 levels and intracellular TG content, and inhibited FFA-induced expression of toll-like receptor 4 (TLR4). In summary, GGQLD abates NASH associated liver injuries via anti-oxidative stress and anti-inflammatory response involved inhibition of TLR4 signal pathways. These findings provide new insights into the anti-NASH therapy by GGQLD.
Collapse
Affiliation(s)
- Chang-Hua Zhang
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Qin Xiao
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Jun-Qing Sheng
- College of Life Science, Nanchang University, Nanchang, 330031, PR China.
| | - Tong-Tong Liu
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ying-Qian Cao
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ya-Nan Xue
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Min Shi
- College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Zheng Cao
- College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Li-Fen Zhou
- Large Precise Instruments Shared Services Center of TCM, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Xiao-Quan Luo
- Experimental Animal Science and Technology Center of TCM, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, PR China
| | - Ke-Zhong Deng
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
37
|
Li Z, Zhao J, Liu H, Wang J, Lu W. Melatonin inhibits apoptosis in mouse Leydig cells via the retinoic acid-related orphan nuclear receptor α/p53 pathway. Life Sci 2020; 246:117431. [PMID: 32061868 DOI: 10.1016/j.lfs.2020.117431] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/07/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
Abstract
Melatonin is an endogenous indoleamine hormone involved in various physiological processes. However, the mechanism of melatonin in mediating Leydig cells function has not been fully explained. In this study, we investigated the mechanism through which melatonin inhibits apoptosis in mouse Leydig cells by activating the retinoic acid-related orphan nuclear receptor (ROR) α/p53 signaling pathway. We confirmed the expression and localization of RORα in mouse Leydig cells using immunofluorescence. After treatment with 10 ng/mL melatonin for 36 h, RORα mRNA and protein levels were significantly increased (P < 0.01). TUNEL and flow cytometry showed that melatonin significantly decreased the TUNEL-positive cell ratio and apoptosis rate (P < 0.05). Moreover, melatonin decreased BAX expression and increased BCL-2 expression (P < 0.05). However, the RORα inhibitor SR1001 reversed the inhibitory effects of melatonin on apoptosis (P < 0.05). Additionally, analysis of p53 expression showed that melatonin inhibited p53 mRNA and protein expression (P < 0.05), whereas SR1001 reversed these effects. p53 reversed the anti-apoptotic process involving RORα-mediated melatonin in mouse Leydig cells. Collectively, our findings suggested that melatonin inhibited apoptosis via the RORα/p53 pathway.
Collapse
Affiliation(s)
- Zhiqiang Li
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Jing Zhao
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Hongyu Liu
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Jun Wang
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China.
| | - Wenfa Lu
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
38
|
Park JS, Moon SJ, Lim MA, Byun JK, Hwang SH, Yang S, Kim EK, Lee H, Kim SM, Lee J, Kwok SK, Min JK, Lee MO, Shin DY, Park SH, Cho ML. Retinoic Acid Receptor-Related Receptor Alpha Ameliorates Autoimmune Arthritis via Inhibiting of Th17 Cells and Osteoclastogenesis. Front Immunol 2019; 10:2270. [PMID: 31636631 PMCID: PMC6787168 DOI: 10.3389/fimmu.2019.02270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/09/2019] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory polyarthritis characterized by progressive joint destruction. IL-17-producing CD4+ T (Th17) cells play pivotal roles in RA development and progression. Retinoic acid receptor-related orphan receptor alpha (RORα) is a negative regulator of inflammatory responses, whereas RORγt, another member of the ROR family, is a Th17 lineage-specific transcription factor. Here, we investigated the immunoregulatory potential of RORα in collagen-induced arthritis (CIA) mice, an experimental model of RA. Cholesterol sulfate (CS) or SR1078, a ligand of RORα, inhibited RORγt expression and Th17 differentiation in vitro. In addition, fortification of RORα in T cells inhibited the expression levels of glycolysis-associated genes. We found that RORα overexpression in CIA mice attenuated the clinical and histological severities of inflammatory arthritis. The anti-arthritic effect of RORα was associated with suppressed Th17 differentiation and attenuated mTOR-STAT3 signaling in T cells. Furthermore, altered RORα activity could directly affect osteoclastogenesis implicated in progressive bone destruction in human RA. Our findings defined a critical role of RORα in the pathogenesis of RA. These data suggest that RORα may have novel therapeutic uses in the treatment of RA.
Collapse
Affiliation(s)
- Jin-Sil Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Su-Jin Moon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Mi-Ae Lim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jae-Kyeong Byun
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sun-Hee Hwang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - SeungCheon Yang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hohyun Lee
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Sung-Min Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jennifer Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jun-Ki Min
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Mi-Ock Lee
- College of Pharmacy and Bio-MAX Institute, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Dong-Yun Shin
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
39
|
RORα suppresses interleukin-6-mediated hepatic acute phase response. Sci Rep 2019; 9:11798. [PMID: 31409825 PMCID: PMC6692401 DOI: 10.1038/s41598-019-48171-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/29/2019] [Indexed: 12/31/2022] Open
Abstract
Acute liver failure (ALF) is characterized by loss of liver function in response to sustained augmentation of the acute-phase response (APR) in the liver, which can progress even to death. Although the inflammatory interleukin-6 (IL-6)–axis is a crucial factor that drives the hepatic APR by releasing diverse acute-phase proteins (APPs), therapeutic strategies to block the IL-6–STAT3-mediated APR are not well developed. Here, we show that the nuclear receptor retinoic acid-related orphan receptor α (RORα) limits APR-mediated liver injury by inhibiting the hepatic IL-6–STAT3 signaling pathway. Administration of JC1-40, an RORα activator, diminished diethylnitrosamine-induced acute liver injury and repressed transcriptional expression of APPs such as CXCL1 and LCN2 in mice. IL-6-mediated activation of STAT3 was repressed after RORα activation by either adenoviral infusion of RORα or JC1-40 treatment in primary hepatocytes. Activation of RORα decreased transcriptional expression of IL-6 receptor α, an upstream activator of STAT3, both in vitro and in vivo. This may be one mechanism underlying the RORα-mediated inhibition of STAT3. Taken together, our results suggest that RORα is a regulator of the hepatic IL-6–STAT3 signaling pathway and may be a new therapeutic target for treating APR-associated inflammatory ALF.
Collapse
|
40
|
Lee DH, Han JH, Lee YS, Jung YS, Roh YS, Yun JS, Han SB, Hong JT. Chitinase-3-like-1 deficiency attenuates ethanol-induced liver injury by inhibition of sterol regulatory element binding protein 1-dependent triglyceride synthesis. Metabolism 2019; 95:46-56. [PMID: 30935969 DOI: 10.1016/j.metabol.2019.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Alcohol overconsumption and abuse lead to alcoholic liver disease (ALD), which is a major chronic liver disease worldwide. Chitinase-3-like protein 1 (CHI3L1) have an important role in the pathogenesis of inflammatory disease. However, the role of CHI3L1 in ALD has not yet been reported. In the present study, we investigated the effect of CHI3L1 on chronic plus binge ethanol-induced liver injury. METHODS CHI3L1 knock out (KO) mice and their littermate control mice based on C57BL/6 (10-12 weeks old) were fed on a Lieber-DeCarli diet containing 6.6% ethanol for 10 days. And, CHI3L1 siRNA or CHI3L1 expressing vector was transfected HepG2 cells were treated with ethanol or without. RESULTS Ethanol-induced hepatic triglyceride (TG) levels and the mRNA levels of TG synthesis-related genes such as acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS) and stearoyl-CoA desaturase-1 (SCD1) were decreased in the liver of CHI3L1 knock out (KO) mice and the HepG2 cells transfected with CHI3L1 siRNA. Increased mRNA level and activation of SREBP1 which is transcription factor of ACC, FAS and SCD1 by ethanol feeding were reduced in the liver of ethanol-fed CHI3L1 KO mice. Moreover, ethanol-induced SREBP1 luciferase activity and mRNA level of SREBP1, ACC, FAS and SCD1 were also decreased in the HepG2 cells transfected with CHI3L1 siRNA, while those were further increased in the HepG2 cells treated with recombinant human CHI3L1. Furthermore, oxidative stress and up-regulated pro-inflammatory cytokines by ethanol were recovered in the liver of ethanol-fed CHI3L1 KO mice. CONCLUSION Our finding suggest that inhibition of CHI3L1 suppressed ethanol-induced liver injury through inhibition of TG synthesis, and the blocking of oxidative stress and hepatic inflammation induced SREBP1 activity could be significant.
Collapse
Affiliation(s)
- Dong Hun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Ji Hye Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Yong Sun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Young Suk Jung
- College of Pharmacy, Pusan National University, 2, Busandaehak-ro 63beon gil, Geumjeong-gu, Busan 609-735, Republic of Korea
| | - Yoon Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jae Suk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Sang Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
41
|
Shi D, Chen J, Wang J, Yao J, Huang Y, Zhang G, Bao Z. Circadian Clock Genes in the Metabolism of Non-alcoholic Fatty Liver Disease. Front Physiol 2019; 10:423. [PMID: 31139087 PMCID: PMC6517678 DOI: 10.3389/fphys.2019.00423] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/27/2019] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common disease, which is characterized by the accumulation of triglycerides in the hepatocytes without excess alcohol intake. Circadian rhythms can participate in lipid, glucose, and cholesterol metabolism and are closely related to metabolism seen in this disease. Circadian clock genes can modulate liver lipid metabolism. Desynchrony of circadian rhythms and the influences imparted by external environmental stimuli can increase morbidity. By contrast, synchronizing circadian rhythms can help to alleviate the metabolic disturbance seen in NAFLD. In this review, we have discussed the current research connections that exist between the circadian clock and the metabolism of NAFLD, and we have specifically focused on the key circadian clock genes, Bmal1, Clock, Rev-Erbs, Rors, Pers, Crys, Nocturnin, and DECs.
Collapse
Affiliation(s)
- Dongmei Shi
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China.,Department of Geriatrics, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Jiaofeng Wang
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China.,Department of Geriatrics, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Jianfeng Yao
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Yiqin Huang
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Gansheng Zhang
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Zhijun Bao
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China.,Department of Geriatrics, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Han S, Li Z, Han F, Jia Y, Qi L, Wu G, Cai W, Xu Y, Li C, Zhang W, Hu D. ROR alpha protects against LPS-induced inflammation by down-regulating SIRT1/NF-kappa B pathway. Arch Biochem Biophys 2019; 668:1-8. [PMID: 31071300 DOI: 10.1016/j.abb.2019.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/15/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023]
Abstract
Systemic inflammatory response syndrome (SIRS) is associated with excessive inflammatory response, however, the pathophysiology of inflammation is poorly understood. The retinoid-related orphan receptor α (RORα) is a key inflammatory regulator, but the mechanisms underlying its role remain unclear. The aim of this study was to investigate how RORα was involved in the regulation of inflammatory response. Here we put forward a hypothesis that RORα might negatively regulate inflammatory response by controlling silent information regulator Sirtuin 1 (SIRT1) expression. Stimulation of macrophages in vitro with LPS and LPS administration in vivo were used to explore the function of RORα and the relationship between RORα and SIRT1. We found that the level of RORα was suppressed in macrophages stimulated with LPS and overexpression or knockdown of RORα by transfection with lentivirus or siRNAs significantly decreased or increased, respectively, the pro-inflammatory cytokines IL-1β, TNF, IL-6 and MCP-1. Importantly, overexpression of RORα suppressed inflammation and alleviated LPS-induced organ injury in vivo. Further study showed that RORα could regulate SIRT1 expression and, consequently, affect deacetyation and nuclear translocation of nuclear factor-kappa B (NF-κB) subunit p65. Moreover, the activation of SIRT1 by its specific agonist, SR1720, could reduce the expression of proinflammatory cytokines in RORα knockdown macrophages stimulated with LPS. In conclusion, we demonstrated that RORα could alleviate LPS-induced inflammation and organ injury both in vivo and in vitro by blocking NF-κB p65 nuclear translocation and restricting acetylation of NF-κB p65 at lysine 310 via the regulation of SIRT1 expression. Targeting RORα might be a promising therapeutic strategy to regulate inflammatory disorders.
Collapse
Affiliation(s)
- Shichao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Zhenzhen Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Fu Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Lijun Qi
- Department of Anesthesiology, Heze Municipal Hospital, Heze, 274031, Shandong, China
| | - Gaofeng Wu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Yongqiang Xu
- Department of Radiology, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Cong Li
- Department of Anesthesiology, Heze Municipal Hospital, Heze, 274031, Shandong, China
| | - Wanfu Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
43
|
Han YH, Shin KO, Kim JY, Khadka DB, Kim HJ, Lee YM, Cho WJ, Cha JY, Lee BJ, Lee MO. A maresin 1/RORα/12-lipoxygenase autoregulatory circuit prevents inflammation and progression of nonalcoholic steatohepatitis. J Clin Invest 2019; 129:1684-1698. [PMID: 30855276 DOI: 10.1172/jci124219] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/24/2019] [Indexed: 12/16/2022] Open
Abstract
Retinoic acid-related orphan receptor α (RORα) is considered a key regulator of polarization in liver macrophages that is closely related to nonalcoholic steatohepatitis (NASH) pathogenesis. However, hepatic microenvironments that support the function of RORα as a polarity regulator were largely unknown. Here, we identified maresin 1 (MaR1), a docosahexaenoic acid (DHA) metabolite with a function of specialized proresolving mediator, as an endogenous ligand of RORα. MaR1 enhanced the expression and transcriptional activity of RORα and thereby increased the M2 polarity of liver macrophages. Administration of MaR1 protected mice from high-fat diet-induced NASH in a RORα-dependent manner. Surprisingly, RORα increased the level of MaR1 through transcriptional induction of 12-lipoxygenase (12-LOX), a key enzyme in MaR1 biosynthesis. Furthermore, we demonstrated that modulation of 12-LOX activity enhanced the protective function of DHA against NASH. Together, these results suggest that the MaR1/RORα/12-LOX autoregulatory circuit could offer potential therapeutic strategies for curing NASH.
Collapse
Affiliation(s)
- Yong-Hyun Han
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Kyong-Oh Shin
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Ju-Yeon Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Daulat B Khadka
- College of Pharmacy, Chonnam National University, Gwangju, South Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Yong-Moon Lee
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Won-Jea Cho
- College of Pharmacy, Chonnam National University, Gwangju, South Korea
| | - Ji-Young Cha
- Laboratory of Cell Metabolism and Gene Regulation, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea
| | - Bong-Jin Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea.,Bio-MAX Institute and.,Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
44
|
Molinaro A, Caesar R, L'homme L, Koh A, Ståhlman M, Staels B, Bäckhed F. Liver-specific RORα deletion does not affect the metabolic susceptibility to western style diet feeding. Mol Metab 2019; 23:82-87. [PMID: 30904385 PMCID: PMC6479759 DOI: 10.1016/j.molmet.2019.02.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
Objectives The nuclear receptor superfamily is a potential target for the development of new treatments for obesity and metabolic diseases. Increasing evidence has pointed towards the retinoic acid-related orphan receptor-alpha (RORα) as an important nuclear receptor involved in several biological processes. RORα full body knockout mice display improved metabolic phenotypes on both chow and high fat (60% fat, 20% carbohydrate) diets, but also have severe behavioral abnormalities. Here we investigated the effect of hepatic RORα by generating mice with liver-specific RORα deletion to elucidate the role of this nuclear receptor on host metabolism. Methods 8 week-old mice with liver-specific RORα deletion and littermate controls were fed either chow or western-style diets (40% fat, 40% carbohydrate) for 12 weeks. Metabolic phenotyping was performed at the end of the dietary intervention. Results Here, we show that hepatic RORα deletion does not affect the metabolic susceptibility to either chow or western-style diet in terms of glucose metabolism and adiposity. Conclusions Our data indicate that liver deletion of RORα does not have a pivotal role in the regulation of hepatic glucose and lipid metabolism on chow or western-style diet. Hepatic deletion of RORα does not affect host metabolism on chow diet. Hepatic deletion of RORα does not affect host metabolism on western-style diet. Similar phenotypes between male and female mice.
Collapse
Affiliation(s)
- Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Robert Caesar
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Laurent L'homme
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000, Lille, France
| | - Ara Koh
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000, Lille, France
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research and Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, 2200, Denmark.
| |
Collapse
|
45
|
Cai J, Jiao X, Fang Y, Yu X, Ding X. The orphan nuclear receptor RORα is a potential endogenous protector in renal ischemia/reperfusion injury. FASEB J 2019; 33:5704-5715. [PMID: 30673513 DOI: 10.1096/fj.201802248r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Emerging evidence indicates that retinoid-related orphan receptor (ROR)α, a member of the ROR nuclear receptor subfamily, mediates key cellular adaptions to hypoxia and contributes to the pathophysiology of many disease states. However, the effects of RORα in renal ischemia/reperfusion (I/R) injury remain unclear. Wild-type (WT) C57 black 6 (C57BL/6) mice and RORα-deficient stagger [ROR(sg/sg)] mice and their WT littermates were used for in vivo studies. The renal I/R injury model was induced by bilateral renal pedicle clamping for 35 min. Human proximal tubule cell line cells were treated with hypoxia (1% oxygen) to establish the cell hypoxia/reoxygenation (H/R) model. We investigated the renal expression and biologic function of RORα, and we found that RORα was significantly down-regulated after renal I/R injury. ROR(sg/sg) mice displayed dramatically augmented renal dysfunction and morphologic damage compared with WT mice at 24 h post-I/R. Further study revealed that the detrimental effects of RORα deficiency were attributable to tubular epithelial cell apoptosis and, consequently, renal inflammation and oxidative stress. The proapoptotic effect of RORα deficiency was associated with aggravated mitochondrial dysfunction in renal tubular cells after I/R. However, pretreatment of C57BL/6 mice with the RORα agonist SR1078 ameliorated I/R-induced renal dysfunction and damage and elicited a concomitant decrease in tubular epithelial cell apoptosis. In summary, our study provides experimental evidence showing that RORα is a novel endogenous protector against renal I/R injury and that ROR-α activation is a promising therapeutic strategy for the prevention of acute kidney injury.-Cai, J., Jiao, X., Fang, Y., Yu, X., Ding, X. The orphan nuclear receptor RORα is a potential endogenous protector in renal ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Jieru Cai
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Shanghai Medical Center for Kidney Disease, Shanghai, China
| | - Xiaoyan Jiao
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Shanghai Medical Center for Kidney Disease, Shanghai, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Shanghai Medical Center for Kidney Disease, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China
| | - Xiaofang Yu
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Shanghai Medical Center for Kidney Disease, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Shanghai Medical Center for Kidney Disease, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China
| |
Collapse
|
46
|
Dornas W, Lagente V. Intestinally derived bacterial products stimulate development of nonalcoholic steatohepatitis. Pharmacol Res 2019; 141:418-428. [PMID: 30658094 DOI: 10.1016/j.phrs.2019.01.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 02/08/2023]
Abstract
Fatty livers are susceptible to factors that cause inflammation and fibrosis, but fat deposition and the inflammatory response can be dissociated. While nonalcoholic fatty liver disease (NAFLD), caused by pathologic fat accumulation inside the liver, can remain stable for several years, in other cases NAFLD progresses to nonalcoholic steatohepatitis (NASH), which is characterized by fat accumulation and inflammation and is not a benign condition. In this review, we discuss the NASH host cells and microbial mechanisms that stimulate inflammation and predispose the liver to hepatocyte injury and fibrotic stages via increased lipid deposition. We highlight the interactions between intestine-derived bacterial products, such as lipopolysaccharide, and nutritional models of NAFLD and/or obese individuals. The results of modulating enteric microbiota suggest that gut-derived endotoxins may be essential determinants of fibrotic progression and regression in NASH.
Collapse
Affiliation(s)
- Waleska Dornas
- NuMeCan Institute (Nutrition, Metabolism and Cancer), Université de Rennes, INSERM, INRA, F-35000 Rennes, France.
| | - Vincent Lagente
- NuMeCan Institute (Nutrition, Metabolism and Cancer), Université de Rennes, INSERM, INRA, F-35000 Rennes, France.
| |
Collapse
|
47
|
Xu Y, Ouyang X, Yan L, Zhang M, Hu Z, Gu J, Fan X, Zhang L, Zhang J, Xue S, Chen G, Su B, Liu J. Sin1 (Stress-Activated Protein Kinase-Interacting Protein) Regulates Ischemia-Induced Microthrombosis Through Integrin αIIbβ3-Mediated Outside-In Signaling and Hypoxia Responses in Platelets. Arterioscler Thromb Vasc Biol 2018; 38:2793-2805. [DOI: 10.1161/atvbaha.118.311822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objective—
Microthrombosis as a serious consequence of myocardial infarction, impairs the microvascular environment and increases the occurrences of heart failure, arrhythmia, and death. Sin1 (stress-activated protein kinase-interacting protein) as an essential component of mTORC2 (mammalian target of rapamycin complex 2) is required for cell proliferation and metabolism in response to nutrients, stress, and reactive oxygen species and activates Akt and PKC (protein kinase C). However, the activation and function of Sin1/mTORC2 in ischemia-induced microthrombosis remain poorly understood.
Approach and Results—
The phosphorylation of the mTORC2 target Akt at S473 (serine 473) was significantly elevated in platelets from the distal end of left anterior descending obstructions from patients who underwent off-pump coronary artery bypass grafting compared with platelets from healthy subjects. Consistent with this finding, phosphorylation of T86 in Sin1 was also dramatically increased. Importantly, the augmented levels of phosphorylated Sin1 and Akt in platelets from 61 preoperative patients with ST-segment—elevation myocardial infarction correlated well with the no-reflow phenomena observed after revascularization. Platelet-specific Sin1 deficiency mice and Sin1 T86 phosphorylation deficiency mice were established to explore the underlying mechanisms in platelet activation. Mechanistically, Sin1 T86 phosphorylation amplifies mTORC2-mediated downstream signals; it is also required for αIIbβ3-mediated outside-in signaling and plays a role in generating hypoxia/reactive oxygen species through NAD
+
/Sirt3 (sirtuin 3)/SOD2 (superoxide dismutase 2) pathway. Importantly, Sin1 deletion in platelets protected mice from ischemia-induced microvascular embolization and subsequent heart dysfunction in a mouse model of myocardial infarction.
Conclusions—
Together, the results of our study reveal a novel role for Sin1 in platelet activation. Thus, Sin1 may be a valuable therapeutic target for interventions for ischemia-induced myocardial infarction deterioration.
Collapse
Affiliation(s)
- Yanyan Xu
- From the Department of Biochemistry and Molecular Cell Biology (Y.X., X.F., L.Z., J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Xinxing Ouyang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology and Molecular Cell Biology (X.O., L.Y., B.S.), Shanghai Jiao Tong University School of Medicine, China
| | - Lichong Yan
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology and Molecular Cell Biology (X.O., L.Y., B.S.), Shanghai Jiao Tong University School of Medicine, China
| | - Mingliang Zhang
- Department of Cardiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China (M.Z., Z.H.)
| | - Zhenlei Hu
- Department of Cardiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China (M.Z., Z.H.)
| | - Jianmin Gu
- Department of Cardiovascular Surgery, Renji Hospital (J.G., S.X.), Shanghai Jiao Tong University School of Medicine, China
| | - Xuemei Fan
- From the Department of Biochemistry and Molecular Cell Biology (Y.X., X.F., L.Z., J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Lin Zhang
- From the Department of Biochemistry and Molecular Cell Biology (Y.X., X.F., L.Z., J.L.), Shanghai Jiao Tong University School of Medicine, China
| | | | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital (J.G., S.X.), Shanghai Jiao Tong University School of Medicine, China
| | - Guoqiang Chen
- Department of Pathophysiology (G.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Bing Su
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology and Molecular Cell Biology (X.O., L.Y., B.S.), Shanghai Jiao Tong University School of Medicine, China
| | - Junling Liu
- From the Department of Biochemistry and Molecular Cell Biology (Y.X., X.F., L.Z., J.L.), Shanghai Jiao Tong University School of Medicine, China
- Collaborative Innovation Center of Hematology, Soochow University, China (J.L.)
| |
Collapse
|
48
|
Dornas W, Glaise D, Bodin A, Sharanek A, Burban A, Le Guillou D, Robert S, Dutertre S, Aninat C, Corlu A, Lagente V. Endotoxin regulates matrix genes increasing reactive oxygen species generation by intercellular communication between palmitate-treated hepatocyte and stellate cell. J Cell Physiol 2018; 234:122-133. [PMID: 30191979 DOI: 10.1002/jcp.27175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/16/2018] [Indexed: 12/19/2022]
Abstract
Previous studies have shown that gut-derived bacterial endotoxins contribute in the progression of simple steatosis to steatohepatitis, although the mechanism(s) remains inaccurate to date. As hepatic stellate cells (HSC) play a pivotal role in the accumulation of excessive extracellular matrix (ECM), leading to collagen deposition, fibrosis, and perpetuation of inflammatory response, an in vitro model was developed to investigate the crosstalk between HSC and hepatocytes (human hepatoma cell) pretreated with palmitate. Bacterial lipopolysaccharide (LPS) stimulated HSC with phosphorylation of the p38 mitogen-activated protein kinase/NF-κB pathway, while several important pro-inflammatory cytokines were upregulated in the presence of hepatocyte-HSC. Concurrently, fibrosis-related genes were regulated by palmitate and the inflammatory effect of endotoxin where cells were more exposed or sensitive to reactive oxygen species (ROS). This interaction was accompanied by increased expression of the mitochondrial master regulator, proliferator-activated receptor gamma coactivator alpha, and a cytoprotective effect of the agent N-acetylcysteine suppressing ROS production, transforming growth factor-β1, and tissue inhibitor of metalloproteinase-1. In summary, our results demonstrate that pro-inflammatory mediators LPS-induced promote ECM rearrangement in hepatic cells transcriptionally committed to the regulation of genes encoding enzymes for fatty acid metabolism in light of differences that might require an alternative therapeutic approach targeting ROS regulation.
Collapse
Affiliation(s)
- Waleska Dornas
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Denise Glaise
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Aude Bodin
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Ahmad Sharanek
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Audrey Burban
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Dounia Le Guillou
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Sacha Robert
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Stephanie Dutertre
- Microscopy Rennes Imaging Center UMS CNRS 3480/US INSERM 018, Biosit, Université de Rennes 1, Rennes, France
| | - Caroline Aninat
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Anne Corlu
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Vincent Lagente
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| |
Collapse
|
49
|
Alshammari GM, Balakrishnan A, Chinnasamy T. Butein protects the nonalcoholic fatty liver through mitochondrial reactive oxygen species attenuation in rats. Biofactors 2018; 44:289-298. [PMID: 29672963 DOI: 10.1002/biof.1428] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/13/2018] [Indexed: 12/13/2022]
Abstract
One of the worldwide metabolic health dilemma is nonalcoholic fatty liver diseases (NAFLD). Researchers are searching effective drug to manage NAFLD patients. One of the best way to manage the metabolic imperfection is through natural principal isolated from different sources. Butein, a natural compound known to have numerous pharmacological application. In the current study we assessed the therapeutic effect of butein administration on liver function tests, oxidative stress, antioxidants, lipid abnormalities, serum inflammatory cytokines, and mitochondrial reactive oxygen species levels, in rats with methionine-choline deficient (MCD) diet induced NAFLD. Male Wistar rats were treated with MCD diet with/without butein (200 mg/kg body wt. orally) for 6 weeks. The protective effect of butein, were evident from decreased transaminase activities, restoration of albumin, globulin, albumin/globulin ratio, and oxidants in serum (P < 0.01), further it improved liver antioxidant status (P < 0.01). Butein significantly lowered lipid profile parameters (P < 0.01), suppressed inflammatory cytokines (P < 0.01), and improved liver histology. Further to understand the possible mechanism behind the hepatoprotective and lipid lowering effect of butein, the activities of heme oxygenase (HO1), myeloperoxidase (MPO), and mitochondrial reactive oxygen species (ROS) were measured. We found that butein supplementation significantly decreased the activity of HO1 (P < 0.001), and increased the activity of MPO (P < 0.001). Furthermore butein attenuated mitochondrial ROS produced in NAFLD condition. Present study shows that butein supplementation restore liver function by altering liver oxidative stress, inflammatory markers, vital defensive enzyme activities, and mitochondrial ROS. In summary, butein has remarkable potential to develop effective hepato-protective drug. © 2018 BioFactors, 44(3):289-298, 2018.
Collapse
Affiliation(s)
- Ghedeir M Alshammari
- Adipocytes and Metabolic Disorders Lab, Food Science and Nutrition Department, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Aristatile Balakrishnan
- Adipocytes and Metabolic Disorders Lab, Food Science and Nutrition Department, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Thirunavukkarasu Chinnasamy
- Adipocytes and Metabolic Disorders Lab, Food Science and Nutrition Department, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
50
|
Han YH, Kim HJ, Na H, Nam MW, Kim JY, Kim JS, Koo SH, Lee MO. RORα Induces KLF4-Mediated M2 Polarization in the Liver Macrophages that Protect against Nonalcoholic Steatohepatitis. Cell Rep 2018; 20:124-135. [PMID: 28683306 DOI: 10.1016/j.celrep.2017.06.017] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 05/04/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022] Open
Abstract
The regulation of M1/M2 polarization in liver macrophages is closely associated with the progression of nonalcoholic steatohepatitis (NASH); however, the mechanism involved in this process remains unclear. Here, we describe the orphan nuclear receptor retinoic-acid-related orphan receptor α (RORα) as a key regulator of M1/M2 polarization in hepatic residential Kupffer cells (KCs) and infiltrated monocyte-derived macrophages. RORα enhanced M2 polarization in KCs by inducing the kruppel-like factor 4. M2 polarization was defective in KCs and bone-marrow-derived macrophages of the myeloid-specific RORα null mice, and these mice were susceptible to HFD-induced NASH. We found that IL-10 played an important role in connecting the function of M2 KCs to lipid accumulation and apoptosis in hepatocytes. Importantly, M2 polarization was controlled by a RORα activator, JC1-40, which improved symptoms of NASH. Our results suggest that the M2-promoting effects of RORα in liver macrophages may provide better therapeutic strategies against NASH.
Collapse
Affiliation(s)
- Yong-Hyun Han
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyelin Na
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Min-Woo Nam
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ju-Yeon Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jun-Seok Kim
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Seung-Hoi Koo
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|