1
|
Zhang F, Cui Y, Zhang T, Yin W. Epigenetic regulation of macrophage activation in chronic obstructive pulmonary disease. Front Immunol 2024; 15:1445372. [PMID: 39206196 PMCID: PMC11349576 DOI: 10.3389/fimmu.2024.1445372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages in the innate immune system play a vital role in various lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury and pulmonary fibrosis. Macrophages involved in the process of immunity need to go through a process of activation, including changes in gene expression and cell metabolism. Epigenetic modifications are key factors of macrophage activation including DNA methylation, histone modification and non-coding RNA regulation. Understanding the role and mechanisms of epigenetic regulation of macrophage activation can provide insights into the function of macrophages in lung diseases and help identification of potential therapeutic targets. This review summarizes the latest progress in the epigenetic changes and regulation of macrophages in their development process and in normal physiological states, and the epigenetic regulation of macrophages in COPD as well as the influence of macrophage activation on COPD development.
Collapse
Affiliation(s)
- Feng Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Yachao Cui
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Tiejun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Fernandes HDB, Oliveira BDS, Machado CA, Carvalho BC, de Brito Toscano EC, da Silva MCM, Vieira ÉLM, de Oliveira ACP, Teixeira AL, de Miranda AS, da Silva AM. Behavioral, neurochemical and neuroimmune features of RasGEF1b deficient mice. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110908. [PMID: 38048936 DOI: 10.1016/j.pnpbp.2023.110908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 11/02/2023] [Accepted: 11/28/2023] [Indexed: 12/06/2023]
Abstract
The factor RasGEF1b is a Ras guanine exchange factor involved in immune responses. Studies have also implicated RasGEF1b in the CNS development. It is still limited the understanding of the role of RasGEF1b in CNS functioning. Using RasGEF1b deficient mice (RasGEF1b-cKO), we investigated the impact of this gene deletion in behavior, cognition, brain neurochemistry and microglia morphology. We showed that RasGEF1b-cKO mice display spontaneous hyperlocomotion and anhedonia. RasGEF1b-cKO mice also exhibited compulsive-like behavior that was restored after acute treatment with the selective serotonin reuptake inhibitor (SSRI) fluoxetine (5 mg/kg). A down-regulation of mRNA of dopamine receptor (Drd1, Drd2, Drd4 and Drd5) and serotonin receptor genes (5Htr1a, 5Htr1b and 5Htr1d) was observed in hippocampus of RasGEF1b-cKO mice. These mice also had reduction of Drd1 and Drd2 in prefrontal cortex and 5Htr1d in striatum. In addition, morphological alterations were observed in RasGEF1b deficient microglia along with decreased levels of hippocampal BDNF. We provided original evidence that the deletion of RasGEF1b leads to unique behavioral features, implicating this factor in CNS functioning.
Collapse
Affiliation(s)
- Heliana de Barros Fernandes
- Laboratório de Genes Inflamatórios, Departamento de Morfologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil; Laboratório de Neurobiologia, Departamento de Morfologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil.
| | - Bruna da Silva Oliveira
- Laboratório de Neurobiologia, Departamento de Morfologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil
| | - Caroline Amaral Machado
- Laboratório de Neurobiologia, Departamento de Morfologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil
| | - Brener Cunha Carvalho
- Laboratório de Genes Inflamatórios, Departamento de Morfologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil
| | - Eliana Cristina de Brito Toscano
- Laboratório Integrado de Pesquisas em Patologia, Departamento de Patologia, Faculdade de Medicina, Universidade Federal de Juiz de Fora, Av. Eugênio do Nascimento, s/n°, Dom Bosco, CEP: 36038-330, Juiz de Fora, MG, Brazil
| | - Maria Carolina M da Silva
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil
| | - Érica Leandro Marciano Vieira
- Campbell Family Mental Health Research Institute, Center of Addiction and Mental Health, 250 College Street, Toronto, ON M5T 1R8, Canada
| | - Antônio Carlos Pinheiro de Oliveira
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Departament of Psychiatry and Behavioral Science McGovern School, Behavioral and Biomedical Sciences Building (BBSB), The University of Texas Health Science Center, 941 East Road, Houston, TX 77054, United States of America
| | - Aline Silva de Miranda
- Laboratório de Neurobiologia, Departamento de Morfologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil
| | - Aristóbolo Mendes da Silva
- Laboratório de Genes Inflamatórios, Departamento de Morfologia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, CEP: 31270-901, Belo Horizonte, MG, Brazil
| |
Collapse
|
3
|
Fernandes HB, de Oliveira IM, Postler TS, Lima SQ, Santos CAC, Oliveira MS, Leão FB, Ghosh S, Souza MC, Andrade W, Silva AM. Transcriptomic analysis reveals that RasGEF1b deletion alters basal and LPS-induced expression of genes involved in chemotaxis and cytokine responses in macrophages. Sci Rep 2023; 13:19614. [PMID: 37950057 PMCID: PMC10638313 DOI: 10.1038/s41598-023-47040-9] [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: 04/25/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023] Open
Abstract
Ras guanine nucleotide exchange factor member 1b (RasGEF1b) of the RasGEF/CDC25 domain-containing family is preferentially expressed by macrophages. However, information is lacking about its role in macrophage function. In this study, we generated mice with ubiquitous deletion of Rasgef1b and used RNA-seq-based transcriptomics to compare the global gene expression in wild-type and knock-out primary bone-marrow-derived macrophages under basal conditions and after lipopolysaccharide (LPS) treatment. Transcriptional filtering identified several genes with significantly different transcript levels between wild-type and knock-out macrophages. In total, 49 and 37 differentially expressed genes were identified at baseline and in LPS-activated macrophages, respectively. Distinct biological processes were significantly linked to down-regulated genes at the basal condition only, and largely included chemotaxis, response to cytokines, and positive regulation of GTPase activity. Importantly, validation by RT-qPCR revealed that the expression of genes identified as down-regulated after LPS stimulation was also decreased in the knock-out cells under basal conditions. We used a luciferase-based reporter assay to showcase the capability of RasGEF1b in activating the Serpinb2 promoter. Notably, knockdown of RasGEF1b in RAW264.7 macrophages resulted in impaired transcriptional activation of the Serpinb2 promoter, both in constitutive and LPS-stimulated conditions. This study provides a small collection of genes that shows relative expression changes effected by the absence of RasGEF1b in macrophages. Thus, we present the first evidence that RasGEF1b mediates the regulation of both steady-state and signal-dependent expression of genes and propose that this GEF plays a role in the maintenance of the basal transcriptional level in macrophages.
Collapse
Affiliation(s)
- Heliana B Fernandes
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Isadora Mafra de Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
- Faculdade de Medicina de Ribeirão Preto, Av. Bandeirantes, 3900 - Campus da USP, Ribeirão Preto, SP, 14049-900, Brazil
| | - Thomas S Postler
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Design and Development Laboratory, International AIDS Vaccine Initiative, Brooklyn, NY, USA
| | - Sérgio Q Lima
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Cícera A C Santos
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia de Rondônia (IFRO), Guajará-Mirim, RO, Brazil
| | - Michaelle S Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Felipe B Leão
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Sankar Ghosh
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Maria C Souza
- Faculdade de Medicina de Ribeirão Preto, Av. Bandeirantes, 3900 - Campus da USP, Ribeirão Preto, SP, 14049-900, Brazil
| | - Warrison Andrade
- Faculdade de Medicina de Ribeirão Preto, Av. Bandeirantes, 3900 - Campus da USP, Ribeirão Preto, SP, 14049-900, Brazil
| | - Aristóbolo M Silva
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
4
|
Penco F, Petretto A, Lavarello C, Papa R, Bertoni A, Omenetti A, Gueli I, Finetti M, Caorsi R, Volpi S, Gattorno M. Proteomic Signatures of Monocytes in Hereditary Recurrent Fevers. Front Immunol 2022; 13:921253. [PMID: 35812440 PMCID: PMC9260596 DOI: 10.3389/fimmu.2022.921253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Hereditary periodic recurrent fevers (HRF) are monogenic autoinflammatory associated to mutations of some genes, such as diseases caused by mutations of including MEFV, TNFRSF1A and MVK genes. Despite the identification of the causative genes, the intracellular implications related to each gene variant are still largely unknown. A large –scale proteomic analysis on monocytes of these patients is aimed to identify with an unbiased approach the mean proteins and molecular interaction networks involved in the pathogenesis of these conditions. Monocytes from HRF 15 patients (5 with MFV, 5 TNFRSF1A and 5with MVK gene mutation) and 15 healthy donors (HDs) were analyzed by liquid chromatography and tandem mass spectrometry before and after lipopolysaccharide (LPS) stimulation. Significant proteins were analyzed through a Cytoscape analysis using the ClueGo app to identify molecular interaction networks. Protein networks for each HRF were performed through a STRING database analysis integrated with a DISEAE database query. About 5000 proteins for each HRF were identified. LPS treatment maximizes differences between up-regulated proteins in monocytes of HRF patients and HDs, independently from the disease’s activity and ongoing treatments. Proteins significantly modulated in monocytes of the different HRF allowed creating a disease-specific proteomic signatures and interactive protein network. Proteomic analysis is able to dissect the different intracellular pathways involved in the inflammatory response of circulating monocytes in HRF patients. The present data may help to identify a “monocyte proteomic signature” for each condition and unravel new possible unexplored intracellular pathways possibly involved in their pathogenesis. These data will be also useful to identify possible differences and similarities between the different HRFs and some multifactorial recurrent fevers.
Collapse
Affiliation(s)
- Federica Penco
- Centro Malattie Autoinfiammatorie ed Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- *Correspondence: Federica Penco, ; Marco Gattorno,
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Chiara Lavarello
- Core Facilities - Clinical Proteomics and Metabolomics, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Riccardo Papa
- Centro Malattie Autoinfiammatorie ed Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Arinna Bertoni
- Centro Malattie Autoinfiammatorie ed Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Alessia Omenetti
- Pediatric Unit, Department of Mother and Child Health, Salesi Children’s Hospital, Ancona, Italy
| | - Ilaria Gueli
- Clinica Pediatrica e Reumatologica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Martina Finetti
- Clinica Pediatrica e Reumatologica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Roberta Caorsi
- Centro Malattie Autoinfiammatorie ed Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Stefano Volpi
- Centro Malattie Autoinfiammatorie ed Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Marco Gattorno
- Centro Malattie Autoinfiammatorie ed Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- *Correspondence: Federica Penco, ; Marco Gattorno,
| |
Collapse
|
5
|
Kumari S, Arora M, Singh J, Kadian LK, Yadav R, Chauhan SS, Chopra A. Molecular Associations and Clinical Significance of RAPs in Hepatocellular Carcinoma. Front Mol Biosci 2021; 8:677979. [PMID: 34235179 PMCID: PMC8255377 DOI: 10.3389/fmolb.2021.677979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive gastrointestinal malignancy with a high rate of mortality. Multiple studies have individually recognized members of RAP gene family as critical regulators of tumor progression in several cancers, including hepatocellular carcinoma. These studies suffer numerous limitations including a small sample size and lack of analysis of various clinicopathological and molecular features. In the current study, we utilized authoritative multi-omics databases to determine the association of RAP gene family expression and detailed molecular and clinicopathological features in hepatocellular carcinoma (HCC). All five RAP genes were observed to harbor dysregulated expression in HCC compared to normal liver tissues. RAP2A exhibited strongest ability to differentiate tumors from the normal tissues. RAP2A expression was associated with progressive tumor grade, TP53 and CTNNB1 mutation status. Additionally, RAP2A expression was associated with the alteration of its copy numbers and DNA methylation. RAP2A also emerged as an independent marker for patient prognosis. Further, pathway analysis revealed that RAP2A expression is correlated with tumor-infiltrating immune cell composition and oncogenic molecular pathways, such as cell cycle and cellular metabolism.
Collapse
Affiliation(s)
- Sarita Kumari
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Mohit Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Jay Singh
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Lokesh K Kadian
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
6
|
Leão FB, Vaughn LS, Bhatt D, Liao W, Maloney D, Carvalho BC, Oliveira L, Ghosh S, Silva AM. Toll-like Receptor (TLR)-induced Rasgef1b expression in macrophages is regulated by NF-κB through its proximal promoter. Int J Biochem Cell Biol 2020; 127:105840. [PMID: 32866686 DOI: 10.1016/j.biocel.2020.105840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/31/2020] [Accepted: 08/21/2020] [Indexed: 12/21/2022]
Abstract
Ras Guanine Exchange Factor (RasGEF) domain family member 1b is encoded by a Toll-like receptor (TLR)-inducible gene expressed in macrophages, but transcriptional mechanisms that govern its expression are still unknown. Here, we have functionally characterized the 5' flanking Rasgef1b sequence and analyzed its transcriptional activation. We have identified that the inflammation-responsive promoter is contained within a short sequence (-183 to +119) surrounding the transcriptional start site. The promoter sequence is evolutionarily conserved and harbors a cluster of five NF-κB binding sites. Luciferase reporter gene assay showed that the promoter is responsive to TLR activation and RelA or cRel, but not RelB, transcription factors. Besides, site-directed mutagenesis showed that the κB binding sites are required for maximal promoter activation induced by LPS. Analysis by Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) revealed that the promoter is located in an accessible chromatin region. More important, Chromatin Immunoprecipitation sequencing (ChIP-seq) showed that RelA is recruited to the promoter region upon LPS stimulation of bone marrow-derived macrophages. Finally, studies with Rela-deficient macrophages or pharmacological inhibition by Bay11-7082 showed that NF-κB is required for optimal Rasgef1b expression induced by TLR agonists. Our data provide evidence of the regulatory mechanism mediated by NF-κB that facilitates Rasgef1b expression after TLR activation in macrophages.
Collapse
Affiliation(s)
- Felipe B Leão
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lauren S Vaughn
- Department of Microbiology & Immunology, Columbia University, College of Physicians and Surgeons, New York, NY 10031, USA
| | - Dev Bhatt
- Department of Microbiology & Immunology, Columbia University, College of Physicians and Surgeons, New York, NY 10031, USA
| | - Will Liao
- New York Genome Center, New York, NY 10013, USA
| | | | - Brener C Carvalho
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Leonardo Oliveira
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sankar Ghosh
- Department of Microbiology & Immunology, Columbia University, College of Physicians and Surgeons, New York, NY 10031, USA
| | - Aristóbolo M Silva
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
7
|
Ren GF, Xiao LL, Ma XJ, Yan YS, Jiao PF. Metformin Decreases Insulin Resistance in Type 1 Diabetes Through Regulating p53 and RAP2A in vitro and in vivo. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2381-2392. [PMID: 32606605 PMCID: PMC7306576 DOI: 10.2147/dddt.s249557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/07/2020] [Indexed: 01/06/2023]
Abstract
Purpose Patients with type 1 diabetes (T1D) are associated with a high risk of multiple complications, so the development of T1D treatment is urgently needed. This study was set out to explore the molecular mechanism of metformin in the treatment of T1D insulin resistance. Patients and Methods Subcutaneous adipose tissues were collected from 68 T1D patients and 51 healthy controls. Insulin resistance model rats and cells were constructed and treated with metformin respectively. Western blot was used to detect p53 and RAP2A protein levels, and qPCR was utilized to measure p53 and RAP2A mRNA levels. SiRNA and RAP2A siRNA vectors were constructed to observe their effects on insulin resistance model cells. Results In T1D, p53 was up-regulated, while RAP2A was down-regulated. Metformin could effectively improve insulin resistance and inflammatory response while down-regulating p53 and up-regulating RAP2A. P53 induced insulin resistance and inflammatory response by inhibiting RAP2A and promoted apoptosis. Conclusion Metformin improves T1D insulin resistance and inflammatory response through p53/RAP2A pathway, and the regulation of p53/RAP2A pathway is conducive to improving the efficacy of metformin in the treatment of insulin resistance.
Collapse
Affiliation(s)
- Gao-Fei Ren
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Li-Li Xiao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Xiao-Jun Ma
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Yu-Shan Yan
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| | - Peng-Fei Jiao
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, People's Republic of China
| |
Collapse
|
8
|
Liu J, Jiang B. Sphk1 promotes ulcerative colitis via activating JAK2/STAT3 signaling pathway. Hum Cell 2019; 33:57-66. [PMID: 31606874 DOI: 10.1007/s13577-019-00283-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/15/2019] [Indexed: 12/11/2022]
Abstract
Ulcerative colitis (UC) is a chronic non-specific inflammatory disease of the colon and rectum. The cause of ulcerative colitis is still unclear, although there may be a hereditary factor. SphK1 has been reported to exhibit an inhibitory effect on the occurrence and development of inflammation; however, the association between SphK1 and the progression of UC remains unclear. The aim of the present study was to investigate the effect of Sphk1 on the progression of UC. The proliferation of RAW264.7 cells was determined using a Cell Counting Kit-8 assay and apoptosis was measured using flow cytometry. The levels of pro-inflammatory cytokines secreted by RAW264.7 cells were investigated using ELISA kits and the protein expression levels in RAW264.7 cells were examined by western blotting. A dextran sulfate sodium (DSS)-induced mouse model was established to investigate the effect of SphK1 on the progression of UC in vivo. Overexpression of Sphk1 significantly increased the proliferation and inhibited the apoptosis of RAW264.7 cells. Additionally, overexpression of Sphk1 increased the secretion of pro-inflammatory cytokines and activated the JAK2/STAT3 signaling pathway in RAW264.7 cells, and JSI-124 partially suppressed these effects. Furthermore, SphK1-small interfering RNA or JSI-124 partially rescued lipopolysaccharide-induced proliferation and pro-inflammatory effects on RAW264.7 cells. The SphK1 inhibitor (PF-543) had an inhibitory effect on DSS-induced UC mice. Sphk1 had significant pro-inflammatory effects on the progression of UC, and may thus be a potential novel therapeutic target for the treatment of UC.
Collapse
Affiliation(s)
- Jiawen Liu
- Department of Gastroenterology, Beijing TsingHua Changgung Hospital, No. 168, LiTang Road, Beijing, 102218, China
| | - Bo Jiang
- Department of Gastroenterology, Beijing TsingHua Changgung Hospital, No. 168, LiTang Road, Beijing, 102218, China.
| |
Collapse
|
9
|
Carvalho BC, Oliveira LC, Rocha CD, Fernandes HB, Oliveira IM, Leãõ FB, Valverde TM, Rego IMG, Ghosh S, Silva AM. Data in support of Rap2a GTPase expression, activation and effects in LPS-mediated innate immune response and NF-κB activation. Data Brief 2019; 24:103965. [PMID: 31193517 PMCID: PMC6531833 DOI: 10.1016/j.dib.2019.103965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/16/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
We present here the data to support the understanding of the implication of Rap2a GTPase in LPS-induced innate immune response and NF-κB activation. The data presented are related to molecular tools that were generated, acquired, optimized or validated to investigate Rap2a expression, activation and its effects in mammalian cells including RAW264.7 macrophages and THP-1 monocytes under inflammatory conditions. These data supplement important technical and biological information on immune function of Rap2a in macrophages activated by LPS, recently reported by us (Carvalho et al., 2019) [1].
Collapse
Affiliation(s)
- Brener C Carvalho
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Leonardo C Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,Department of Microbiology & Immunology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Carolina D Rocha
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Heliana B Fernandes
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Isadora M Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Felipe B Leãõ
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Thalita M Valverde
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Igor M G Rego
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sankar Ghosh
- Department of Microbiology & Immunology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Aristóbolo M Silva
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|