101
|
Sun C, Wu G, Zhang Z, Cao R, Cui S. Protein Tyrosine Phosphatase Receptor Type D Regulates Neuropathic Pain After Nerve Injury via the STING-IFN-I Pathway. Front Mol Neurosci 2022; 15:859166. [PMID: 35493326 PMCID: PMC9047945 DOI: 10.3389/fnmol.2022.859166] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
Neuropathic pain is usually caused by injury or dysfunction of the somatosensory system, and medicine is a common way of treatment. Currently, there are still no satisfactory drugs, like opioids and lidocaine, which carry a high risk of addiction. Protein tyrosine phosphatase receptor type D (PTPRD) is a known therapeutic target in addiction pathways and small molecule inhibitors targeting it, such as 7-butoxy illudalic acid analog (7-BIA), have recently been developed to tackle addition. PTPRD is also upregulated in the dorsal root ganglion (DRG) in a rat model of neuropathic pain, but is not yet clear whether PTPRD contributes to the development of neuropathic pain. Here, we established a chronic constriction injury (CCI) and evaluated PTPRD expression and its association with neuropathic pain. PTPRD expression was found to gradually increase after CCI in DRGs, and its expression was concomitant with the progressive development of hypersensitivity as assessed by both mechanical and thermal stimuli. Both PTPRD knockdown and administration of PTPRD inhibitor 7-BIA alleviated CCI-induced neuropathic pain while upregulating STING and IFN-α in the DRG. Treatment with H-151, a STING inhibitor, abolished the analgesic effects of PTPRD knockdown. Taken together, our study suggests that increased levels of PTPRD in the DRG following CCI are involved in the development of neuropathic pain via the STING-IFN-I pathway. 7-BIA, a small molecule inhibitor of PTPRD with anti-addiction effects, may represent a novel and safe therapeutic strategy for the clinical management of neuropathic pain without the risk of addiction.
Collapse
|
102
|
An overview of the crosstalk between YAP and cGAS-STING signaling in non-small cell lung cancer: it takes two to tango. Clin Transl Oncol 2022; 24:1661-1672. [PMID: 35377059 DOI: 10.1007/s12094-022-02826-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is recognized as a main mediator bridging innate and adaptive immunity, recent advances have expanded its roles to anti-tumor immunity and carcinogenesis. Loss of cGAS-STING signaling in non-small cell lung cancer (NSCLC) leads to enhanced tumorigenicity and decreased cytotoxic T lymphocyte infiltration. Apart from its anticancer response, persistent overreaction of cGAS-STING signaling promotes progression of certain inflammation-aggravated cancers. Activation of the pro-inflammatory nucleic acid sensing pathway can trigger Hippo pathway, which mediates the inactivation of Yes-associated protein 1 (YAP1) and its paralogue transcriptional co-regulators with PDZ-binding motif (TAZ, also known as WWTR1), and subsequent suppression of tumorigenesis. Active YAP acts as a transcriptional driver in bolstering immunosuppressive cytokines to evade immune surveillance and promote occurrence of preneoplasia. It is reasonable that aggressive tumors co-opt these regulators to generate few immunogenic antigens and drive tumorigenic behaviors via a highly cooperative manner. Given their multifaced roles, we profile the molecular biology characteristic and current status underpinning oncogenic YAP, review its crosstalk roles with cGAS/STING pathway in NSCLC, and summarize the major clinical investigations in NSCLC with TCGA database.
Collapse
|
103
|
Patel SJ, Liu N, Piaker S, Gulko A, Andrade ML, Heyward FD, Sermersheim T, Edinger N, Srinivasan H, Emont MP, Westcott GP, Luther J, Chung RT, Yan S, Kumari M, Thomas R, Deleye Y, Tchernof A, White PJ, Baselli GA, Meroni M, De Jesus DF, Ahmad R, Kulkarni RN, Valenti L, Tsai L, Rosen ED. Hepatic IRF3 fuels dysglycemia in obesity through direct regulation of Ppp2r1b. Sci Transl Med 2022; 14:eabh3831. [PMID: 35320000 PMCID: PMC9162056 DOI: 10.1126/scitranslmed.abh3831] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammation has profound but poorly understood effects on metabolism, especially in the context of obesity and nonalcoholic fatty liver disease (NAFLD). Here, we report that hepatic interferon regulatory factor 3 (IRF3) is a direct transcriptional regulator of glucose homeostasis through induction of Ppp2r1b, a component of serine/threonine phosphatase PP2A, and subsequent suppression of glucose production. Global ablation of IRF3 in mice on a high-fat diet protected against both steatosis and dysglycemia, whereas hepatocyte-specific loss of IRF3 affects only dysglycemia. Integration of the IRF3-dependent transcriptome and cistrome in mouse hepatocytes identifies Ppp2r1b as a direct IRF3 target responsible for mediating its metabolic actions on glucose homeostasis. IRF3-mediated induction of Ppp2r1b amplified PP2A activity, with subsequent dephosphorylation of AMPKα and AKT. Furthermore, suppression of hepatic Irf3 expression with antisense oligonucleotides reversed obesity-induced insulin resistance and restored glucose homeostasis in obese mice. Obese humans with NAFLD displayed enhanced activation of liver IRF3, with reversion after bariatric surgery. Hepatic PPP2R1B expression correlated with HgbA1C and was elevated in obese humans with impaired fasting glucose. We therefore identify the hepatic IRF3-PPP2R1B axis as a causal link between obesity-induced inflammation and dysglycemia and suggest an approach for limiting the metabolic dysfunction accompanying obesity-associated NAFLD.
Collapse
Affiliation(s)
- Suraj J. Patel
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Digestive and Liver Diseases, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nan Liu
- Harvard Medical School, Boston, MA 02115, USA
- Cancer and Blood Disorders Center, Dana Farber Cancer Institute and Boston Children’s Hospital, Boston, MA 02215, USA
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| | - Sam Piaker
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Anton Gulko
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Maynara L. Andrade
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Frankie D. Heyward
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Tyler Sermersheim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Nufar Edinger
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Harini Srinivasan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Margo P. Emont
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Gregory P. Westcott
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jay Luther
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Raymond T. Chung
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Shuai Yan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Manju Kumari
- Department of Cardiology, Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Reeby Thomas
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Yann Deleye
- Duke Molecular Physiology Institute and Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - André Tchernof
- Institut Universitaire de Cardiologie and Pneumologie de Québec–Université Laval (IUCPQUL), Québec City, Canada
| | - Phillip J. White
- Duke Molecular Physiology Institute and Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido A. Baselli
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario F. De Jesus
- Harvard Medical School, Boston, MA 02115, USA
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rohit N. Kulkarni
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linus Tsai
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Evan D. Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
104
|
Chou WC, Rampanelli E, Li X, Ting JPY. Impact of intracellular innate immune receptors on immunometabolism. Cell Mol Immunol 2022; 19:337-351. [PMID: 34697412 PMCID: PMC8891342 DOI: 10.1038/s41423-021-00780-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/17/2021] [Indexed: 12/21/2022] Open
Abstract
Immunometabolism, which is the metabolic reprogramming of anaerobic glycolysis, oxidative phosphorylation, and metabolite synthesis upon immune cell activation, has gained importance as a regulator of the homeostasis, activation, proliferation, and differentiation of innate and adaptive immune cell subsets that function as key factors in immunity. Metabolic changes in epithelial and other stromal cells in response to different stimulatory signals are also crucial in infection, inflammation, cancer, autoimmune diseases, and metabolic disorders. The crosstalk between the PI3K-AKT-mTOR and LKB1-AMPK signaling pathways is critical for modulating both immune and nonimmune cell metabolism. The bidirectional interaction between immune cells and metabolism is a topic of intense study. Toll-like receptors (TLRs), cytokine receptors, and T and B cell receptors have been shown to activate multiple downstream metabolic pathways. However, how intracellular innate immune sensors/receptors intersect with metabolic pathways is less well understood. The goal of this review is to examine the link between immunometabolism and the functions of several intracellular innate immune sensors or receptors, such as nucleotide-binding and leucine-rich repeat-containing receptors (NLRs, or NOD-like receptors), absent in melanoma 2 (AIM2)-like receptors (ALRs), and the cyclic dinucleotide receptor stimulator of interferon genes (STING). We will focus on recent advances and describe the impact of these intracellular innate immune receptors on multiple metabolic pathways. Whenever appropriate, this review will provide a brief contextual connection to pathogenic infections, autoimmune diseases, cancers, metabolic disorders, and/or inflammatory bowel diseases.
Collapse
Affiliation(s)
- Wei-Chun Chou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Elena Rampanelli
- Amsterdam UMC (University Medical Center, location AMC), Department of Experimental Vascular Medicine, AGEM (Amsterdam Gastroenterology Endocrinology Metabolism) Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Xin Li
- Comparative Immunology Research Center, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
105
|
A distinct role of STING in regulating glucose homeostasis through insulin sensitivity and insulin secretion. Proc Natl Acad Sci U S A 2022; 119:2101848119. [PMID: 35145023 PMCID: PMC8851542 DOI: 10.1073/pnas.2101848119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
The role of STING in maintaining glucose homeostasis remains unknown. Herein, using global and β-cell–specific STING knockout mouse models, we revealed a distinct role of STING in the regulation of glucose homeostasis through β-cells and peripheral tissues. Specially, while global STING knockout beneficially alleviated insulin resistance and glucose intolerance induced by high-fat diet, it surprisingly impaired islet glucose-stimulated insulin secretion (GSIS). Further analyses revealed that STING deficiency down-regulated expression of β-cell key transcription factor Pax6, impairing Pax6 nuclear localization and binding activity to the promoters of its target genes, including Glut2 and Abcc8, causing impaired GSIS. These data highlight pathophysiological significance of fine-tuned STING signaling in β-cells and insulin target tissues for maintaining glucose homeostasis. Insulin resistance and β-cell dysfunction are two main molecular bases yet to be further elucidated for type 2 diabetes (T2D). Accumulating evidence indicates that stimulator of interferon genes (STING) plays an important role in regulating insulin sensitivity. However, its function in β-cells remains unknown. Herein, using global STING knockout (STING−/−) and β-cell–specific STING knockout (STING-βKO) mouse models, we revealed a distinct role of STING in the regulation of glucose homeostasis through peripheral tissues and β-cells. Specially, although STING−/− beneficially alleviated insulin resistance and glucose intolerance induced by high-fat diet, it surprisingly impaired islet glucose-stimulated insulin secretion (GSIS). Importantly, STING is decreased in islets of db/db mice and patients with T2D, suggesting a possible role of STING in β-cell dysfunction. Indeed, STING-βKO caused glucose intolerance due to impaired GSIS, indicating that STING is required for normal β-cell function. Islet transcriptome analysis showed that STING deficiency decreased expression of β-cell function–related genes, including Glut2, Kcnj11, and Abcc8, contributing to impaired GSIS. Mechanistically, the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and cleavage under targets and tagmentation (CUT&Tag) analyses suggested that Pax6 was the transcription factor that might be associated with defective GSIS in STING-βKO mice. Indeed, Pax6 messenger RNA and protein levels were down-regulated and its nuclear localization was lost in STING-βKO β-cells. Together, these data revealed a function of STING in the regulation of insulin secretion and established pathophysiological significance of fine-tuned STING within β-cells and insulin target tissues for maintaining glucose homeostasis.
Collapse
|
106
|
Liu K, Jin X, Zhang X, Lian H, Ye J. The mechanisms of nucleotide actions in insulin resistance. J Genet Genomics 2022; 49:299-307. [DOI: 10.1016/j.jgg.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/14/2022]
|
107
|
Bryant JD, Lei Y, VanPortfliet JJ, Winters AD, West AP. Assessing Mitochondrial DNA Release into the Cytosol and Subsequent Activation of Innate Immune-related Pathways in Mammalian Cells. Curr Protoc 2022; 2:e372. [PMID: 35175686 PMCID: PMC8986093 DOI: 10.1002/cpz1.372] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mitochondria have emerged as key drivers of mammalian innate immune responses, functioning as signaling hubs to trigger inflammation and orchestrating metabolic switches required for phagocyte activation. Mitochondria also contain damage-associated molecular patterns (DAMPs), molecules that share similarity with pathogen-associated molecular patterns (PAMPs) and can engage innate immune sensors to drive inflammation. The aberrant release of mitochondrial DAMPs during cellular stress and injury is an increasingly recognized trigger of inflammatory responses in human diseases. Mitochondrial DNA (mtDNA) is a particularly potent DAMP that engages multiple innate immune sensors, although mounting evidence suggests that cytosolic mtDNA is primarily detected via the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. cGAS and STING are widely expressed in mammalian cells and serve as key regulators of type I interferon and cytokine expression in both infectious and inflammatory diseases. Despite growing roles for the mtDNA-cGAS-STING axis in human disease, assays to quantify mtDNA release into the cytosol and approaches to link mtDNA to cGAS-STING signaling are not standardized, which increases the possibility for experimental artifacts and misinterpretation of data. Here, we present a series of protocols for assaying the release of mtDNA into the cytosol and subsequent activation of innate immune signaling in mammalian cells. We highlight genetic and pharmacological approaches to induce and inhibit mtDNA release from mitochondria. We also describe immunofluorescence microscopy and cellular fractionation assays to visualize morphological changes in mtDNA and quantify mtDNA accumulation in the cytosol. Finally, we include protocols to examine mtDNA-dependent cGAS-STING activation by RT-qPCR and western blotting. These methods can be performed with standard laboratory equipment and are highly adaptable to a wide range of mammalian cell types. They will permit researchers working across the spectrum of biological and biomedical sciences to accurately and reproducibly measure cytosolic mtDNA release and resulting innate immune responses. © 2022 Wiley Periodicals LLC. Basic Protocol 1: siRNA-mediated knockdown of TFAM to induce mtDNA instability, cytosolic release, and activation of the cGAS-STING pathway Alternate Protocol: Pharmacological induction of mtDNA release and cGAS-STING activation using ABT-737 and Q-VD-OPH Basic Protocol 2: Isolation and quantitation of DNA from cytosolic, mitochondrial, and nuclear fractions Basic Protocol 3: Pharmacological inhibition of mtDNA replication and release.
Collapse
Affiliation(s)
- Joshua D. Bryant
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
- These authors contributed equally
| | - Yuanjiu Lei
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
- These authors contributed equally
| | - Jordyn J. VanPortfliet
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
| | - Ashley D. Winters
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
| | - A. Phillip West
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
| |
Collapse
|
108
|
Cao L, Xu E, Zheng R, Zhangchen Z, Zhong R, Huang F, Ye J, Sun H, Fan Y, Xie S, Chen Y, Xu Y, Cao J, Cao W, Liu C. Traditional Chinese medicine Lingguizhugan decoction ameliorate HFD-induced hepatic-lipid deposition in mice by inhibiting STING-mediated inflammation in macrophages. Chin Med 2022; 17:7. [PMID: 34983596 PMCID: PMC8728979 DOI: 10.1186/s13020-021-00559-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/18/2021] [Indexed: 11/25/2022] Open
Abstract
Background Stimulator of IFN genes (STING) is highly expressed in the livers of non-alcoholic fatty liver disease (NAFLD) patients and high fat diet (HFD) induced NAFLD mice model. The STING signaling-mediated inflammation has been shown to play a critical role in metabolic disorders. Lingguizhugan decoction (LGZG), a Traditional Chinese herbal decoction, has been applied to treat metabolic disorders for many years. However, whether LGZG can alleviate the progression of NAFLD through inhibiting inflammation remains unclear. This study was to determine the role of STING-mediated inflammation in the HFD-induced hepatic-lipid deposition treated with LGZG. Methods The anti-inflammatory and anti-steatotic effects of LGZG in vivo were detected by H&E staining, immunofluorescence and immuno-chemistry. Mice bone-marrow-derived macrophages (BMDMs) and primary liver macrophages were treated with STING-specific agonist (DMXAA), LGZG and its critical components respectively. The treated culture supernatant of BMDMs and primary liver macrophages from each group was co-cultured with palmitic acid-treated mouse primary hepatocytes or mouse liver cell line AML-12 respectively to detect whether the activation of STING-mediated pathway is involved in the anti-steatotic effect of LGZG. The hepatocyte lipid deposition in vivo and in vitro were detected by oil red staining. Mitochondrial DNA release of mouse liver extracts were detected by real time PCR. The expression of proteins and inflammatory cytokines related to STING-TBK1-NF-κB pathway was detected by western blotting and ELISA. Results LGZG significantly ameliorated HFD induced hepatic steatosis, oxidative stress, hepatic mitochondrial damage and mitochondrial DNA release, which was correlated with reduction of the expression level of STING as well as the infiltration of STING-positive macrophages in the livers of HFD fed mice. The critical components of LGZG directly inhibited the activation of STING-TBK1-NF-κB pathway in liver macrophages induced by DMXAA, LPS, thereby reducing the release of IFNβ and TNFα. Co-incubating the culture supernatant of LGZG treated liver macrophages and PA-stimulated hepatocytes significantly inhibited the PA-induced lipid deposition. Conclusion This study demonstrates that LGZG can ameliorate HFD-induced hepatic-lipid deposition through inhibiting STING-TBK1-NF-κB pathway in liver macrophages, which provides novel insight for elucidating the molecular mechanism of LGZG alleviating HFD induced hepatic steatosis.
Collapse
Affiliation(s)
- Lin Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.
| | - Erjin Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Rendong Zheng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Zhili Zhangchen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Rongling Zhong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Fei Huang
- Suzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No. 18 Yangsu Road, Gusu District, Suzhou, 215002, China
| | - Juan Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Hongping Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Yaofu Fan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Shaofeng Xie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Yu Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Yijiao Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China
| | - Jing Cao
- Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Qinhuai District, Nanjing, 210029, China
| | - Wen Cao
- The Affiliated Jiangning Hospital of Nanjing Medical University, No. 169, Dongshan street, Hushan Road, Jiangning District, Nanjing, 211100, China.
| | - Chao Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, No. 100, Shizi Street, Hongshan Road, Nanjing, 210028, China.
| |
Collapse
|
109
|
Liu H, Wang F, Cao Y, Dang Y, Ge B. OUP accepted manuscript. J Mol Cell Biol 2022; 14:6583286. [PMID: 35536585 PMCID: PMC9475664 DOI: 10.1093/jmcb/mjac031] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/14/2022] [Accepted: 05/06/2022] [Indexed: 11/12/2022] Open
Abstract
Pattern recognition receptors are critical for the sensing of pathogen-associated molecular patterns or danger-associated molecular patterns and subsequent mounting of innate immunity and shaping of adaptive immunity. The identification of 2′3′-cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) synthase (cGAS) as a major cytosolic DNA receptor is a milestone in the field of DNA sensing. The engagement of cGAS by double-stranded DNA from different origins, including invading pathogens, damaged mitochondria, ruptured micronuclei, and genomic DNA results in the generation of cGAMP and activation of stimulator of interferon genes, which thereby activates innate immunity mainly characterized by the activation of type I interferon response. In recent years, great progress has been made in understanding the subcellular localization and novel functions of cGAS. In this review, we particularly focus on summarizing the multifaceted roles of cGAS in regulating senescence, autophagy, cell stemness, apoptosis, angiogenesis, cell proliferation, antitumor effect, DNA replication, DNA damage repair, micronucleophagy, as well as cell metabolism.
Collapse
Affiliation(s)
| | - Fei Wang
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yajuan Cao
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yifang Dang
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Baoxue Ge
- Correspondence to: Baoxue Ge, E-mail:
| |
Collapse
|
110
|
The cGAS-STING signaling in cardiovascular and metabolic diseases: Future novel target option for pharmacotherapy. Acta Pharm Sin B 2022; 12:50-75. [PMID: 35127372 PMCID: PMC8799861 DOI: 10.1016/j.apsb.2021.05.011] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/05/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling exert essential regulatory function in microbial-and onco-immunology through the induction of cytokines, primarily type I interferons. Recently, the aberrant and deranged signaling of the cGAS-STING axis is closely implicated in multiple sterile inflammatory diseases, including heart failure, myocardial infarction, cardiac hypertrophy, nonalcoholic fatty liver diseases, aortic aneurysm and dissection, obesity, etc. This is because of the massive loads of damage-associated molecular patterns (mitochondrial DNA, DNA in extracellular vesicles) liberated from recurrent injury to metabolic cellular organelles and tissues, which are sensed by the pathway. Also, the cGAS-STING pathway crosstalk with essential intracellular homeostasis processes like apoptosis, autophagy, and regulate cellular metabolism. Targeting derailed STING signaling has become necessary for chronic inflammatory diseases. Meanwhile, excessive type I interferons signaling impact on cardiovascular and metabolic health remain entirely elusive. In this review, we summarize the intimate connection between the cGAS-STING pathway and cardiovascular and metabolic disorders. We also discuss some potential small molecule inhibitors for the pathway. This review provides insight to stimulate interest in and support future research into understanding this signaling axis in cardiovascular and metabolic tissues and diseases.
Collapse
Key Words
- AA, amino acids
- AAD, aortic aneurysm and dissection
- AKT, protein kinase B
- AMPK, AMP-activated protein kinase
- ATP, adenosine triphosphate
- Ang II, angiotensin II
- CBD, C-binding domain
- CDG, c-di-GMP
- CDNs, cyclic dinucleotides
- CTD, C-terminal domain
- CTT, C-terminal tail
- CVDs, cardiovascular diseases
- Cardiovascular diseases
- Cys, cysteine
- DAMPs, danger-associated molecular patterns
- Damage-associated molecular patterns
- DsbA-L, disulfide-bond A oxidoreductase-like protein
- ER stress
- ER, endoplasmic reticulum
- GTP, guanosine triphosphate
- HAQ, R71H-G230A-R293Q
- HFD, high-fat diet
- ICAM-1, intracellular adhesion molecule 1
- IFN, interferon
- IFN-I, type 1 interferon
- IFNAR, interferon receptors
- IFNIC, interferon-inducible cells
- IKK, IκB kinase
- IL, interleukin
- IRF3, interferon regulatory factor 3
- ISGs, IRF-3-dependent interferon-stimulated genes
- Inflammation
- LBD, ligand-binding pocket
- LPS, lipopolysaccharides
- MI, myocardial infarction
- MLKL, mixed lineage kinase domain-like protein
- MST1, mammalian Ste20-like kinases 1
- Metabolic diseases
- Mitochondria
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor-kappa B
- NLRP3, NOD-, LRR- and pyrin domain-containing protein 3
- NO2-FA, nitro-fatty acids
- NTase, nucleotidyltransferase
- PDE3B/4, phosphodiesterase-3B/4
- PKA, protein kinase A
- PPI, protein–protein interface
- Poly: I.C, polyinosinic-polycytidylic acid
- ROS, reactive oxygen species
- SAVI, STING-associated vasculopathy with onset in infancy
- SNPs, single nucleotide polymorphisms
- STIM1, stromal interaction molecule 1
- STING
- STING, stimulator of interferon genes
- Ser, serine
- TAK1, transforming growth factor β-activated kinase 1
- TBK1, TANK-binding kinase 1
- TFAM, mitochondrial transcription factor A
- TLR, Toll-like receptors
- TM, transmembrane
- TNFα, tumor necrosis factor-alpha
- TRAF6, tumor necrosis factor receptor-associated factor 6
- TREX1, three prime repair exonuclease 1
- YAP1, Yes-associated protein 1
- cGAMP, 2′,3′-cyclic GMP–AMP
- cGAS
- cGAS, cyclic GMP–AMP synthase
- dsDNA, double-stranded DNA
- hSTING, human stimulator of interferon genes
- mTOR, mammalian target of rapamycin
- mtDNA, mitochondrial DNA
Collapse
|
111
|
Wu JT, He BW, Cao JL, Yan JB, Chen ZY. Involvement of STING signaling pathway in non-alcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2021; 29:1396-1401. [DOI: 10.11569/wcjd.v29.i24.1396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the major chronic liver diseases worldwide, which seriously threatens human health and has become a major public health problem. Immune mechanism plays a key role in the occurrence and development of NAFLD. Interferon gene stimulating factor (STING) is a key connector protein of the immune system, and its related signaling pathway has become a recent hot research topic. This signaling pathway may be involved in the occurrence and development of NAFLD by mediating liver inflammation, lipid metabolism, apoptosis, and other processes that affect liver metabolic homeostasis. Combining relevant reports and the latest literature, this paper reviews NAFLD and immunity, the composition of STING signaling pathway, and the relationship between the STING signaling pathway and NAFLD, in order to provide ideas for further in-depth study of the complex relationship between the STING signaling pathway and NAFLD and the development of relevant targeted drugs.
Collapse
Affiliation(s)
- Jin-Ting Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou Command, Hangzhou 310006, Zhejiang Province, China
| | - Bo-Wu He
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou Command, Hangzhou 310006, Zhejiang Province, China
| | - Jie-Lu Cao
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou Command, Hangzhou 310006, Zhejiang Province, China
| | - Jun-Bin Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou Command, Hangzhou 310006, Zhejiang Province, China
| | - Zhi-Yun Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou Command, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
112
|
Bao T, Liu J, Leng J, Cai L. The cGAS-STING pathway: more than fighting against viruses and cancer. Cell Biosci 2021; 11:209. [PMID: 34906241 PMCID: PMC8670263 DOI: 10.1186/s13578-021-00724-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 12/02/2021] [Indexed: 01/07/2023] Open
Abstract
In the classic Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway, downstream signals can control the production of type I interferon and nuclear factor kappa-light-chain-enhancer of activated B cells to promote the activation of pro-inflammatory molecules, which are mainly induced during antiviral responses. However, with progress in this area of research, studies focused on autoimmune diseases and chronic inflammatory conditions that may be relevant to cGAS-STING pathways have been conducted. This review mainly highlights the functions of the cGAS-STING pathway in chronic inflammatory diseases. Importantly, the cGAS-STING pathway has a major impact on lipid metabolism. Different research groups have confirmed that the cGAS-STING pathway plays an important role in the chronic inflammatory status in various organs. However, this pathway has not been studied in depth in diabetes and diabetes-related complications. Current research on the cGAS-STING pathway has shown that the targeted therapy of diseases that may be caused by inflammation via the cGAS-STING pathway has promising outcomes.
Collapse
Affiliation(s)
- Terigen Bao
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
- Department of Pediatrics, The Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Jia Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jiyan Leng
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Lu Cai
- Department of Pediatrics, The Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, 40292, USA
- Departments of Pharmacology and Toxicology, The University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
113
|
Wang L, You HM, Meng HW, Pan XY, Chen X, Bi YH, Zhang YF, Li JJ, Yin NN, Zhang ZW, Huang C, Li J. STING-mediated inflammation contributes to Gao binge ethanol feeding model. J Cell Physiol 2021; 237:1471-1485. [PMID: 34698390 PMCID: PMC9298121 DOI: 10.1002/jcp.30606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/14/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
Alcohol metabolism causes hepatocytes to release damage‐associated molecular patterns (DAMPs). This includes mitochondrial DNA (mtDNA), which is generated and released from damaged hepatocytes and contributes to liver injury by producing proinflammatory cytokines. STING is a pattern recognition receptor of DAMPs known to control the induction of innate immunity in various pathological processes. However, the expression profile and functions of STING in the Gao binge ethanol model remain poorly understood. We demonstrated that STING is upregulated in the Gao binge ethanol model. STING functions as an mtDNA sensor in the Kupffer cells of the liver and induces STING‐signaling pathway‐dependent inflammation and further aggravates hepatocyte apoptosis in the Gao binge ethanol model. This study provides novel insights into predicting disease progression and developing targeted therapies for alcoholic liver injury.
Collapse
Affiliation(s)
- Ling Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hong-Mei You
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hong-Wu Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xue-Yin Pan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xin Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yi-Hui Bi
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ya-Fei Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan-Juan Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Na-Na Yin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Zheng-Wei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
114
|
Visitchanakun P, Kaewduangduen W, Chareonsappakit A, Susantitaphong P, Pisitkun P, Ritprajak P, Townamchai N, Leelahavanichkul A. Interference on Cytosolic DNA Activation Attenuates Sepsis Severity: Experiments on Cyclic GMP-AMP Synthase (cGAS) Deficient Mice. Int J Mol Sci 2021; 22:ijms222111450. [PMID: 34768881 PMCID: PMC8583992 DOI: 10.3390/ijms222111450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
Although the enhanced responses against serum cell-free DNA (cfDNA) in cases of sepsis—a life-threatening organ dysfunction due to systemic infection—are understood, the influence of the cytosolic DNA receptor cGAS (cyclic guanosine monophosphate–adenosine monophosphate (GMP–AMP) synthase) on sepsis is still unclear. Here, experiments on cGAS deficient (cGAS-/-) mice were conducted using cecal ligation and puncture (CLP) and lipopolysaccharide (LPS) injection sepsis models and macrophages. Severity of CLP in cGAS-/- mice was less severe than in wildtype (WT) mice, as indicated by mortality, serum LPS, cfDNA, leukopenia, cytokines (TNF-α, IL-6 and IL-10), organ histology (lung, liver and kidney) and spleen apoptosis. With the LPS injection model, serum cytokines in cGAS-/- mice were lower than in WT mice, despite the similar serum cfDNA level. Likewise, in LPS-activated WT macrophages, the expression of several mitochondria-associated genes (as revealed by RNA sequencing analysis) and a profound reduction in mitochondrial parameters, including maximal respiration (determined by extracellular flux analysis), DNA (mtDNA) and mitochondrial abundance (revealed by fluorescent staining), were demonstrated. These data implied the impact of cfDNA resulting from LPS-induced cell injury. In parallel, an additive effect of bacterial DNA on LPS, seen in comparison with LPS alone, was demonstrated in WT macrophages, but not in cGAS-/- cells, as indicated by supernatant cytokines (TNF-α and IL-6), M1 proinflammatory polarization (iNOS and IL-1β), cGAS, IFN-γ and supernatant cyclic GMP–AMP (cGAMP). In conclusion, cGAS activation by cfDNA from hosts (especially mtDNA) and bacteria was found to induce an additive proinflammatory effect on LPS-activated macrophages which was perhaps responsible for the more pronounced sepsis hyperinflammation observed in WT mice compared with the cGAS-/- group.
Collapse
Affiliation(s)
- Peerapat Visitchanakun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.V.); (W.K.); (A.C.)
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Warerat Kaewduangduen
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.V.); (W.K.); (A.C.)
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Awirut Chareonsappakit
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.V.); (W.K.); (A.C.)
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Paweena Susantitaphong
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Research Unit for Metabolic Bone Disease in CKD Patients, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10330, Thailand;
| | - Patcharee Ritprajak
- Research Unit in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Natavudh Townamchai
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (N.T.); (A.L.); Tel.: +66-2-256-4251 (N.T. & A.L.); Fax: +66-2-252-6920 (N.T. & A.L.)
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.V.); (W.K.); (A.C.)
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (N.T.); (A.L.); Tel.: +66-2-256-4251 (N.T. & A.L.); Fax: +66-2-252-6920 (N.T. & A.L.)
| |
Collapse
|
115
|
Bai J, Liu F. cGAS‒STING signaling and function in metabolism and kidney diseases. J Mol Cell Biol 2021; 13:728-738. [PMID: 34665236 PMCID: PMC8718186 DOI: 10.1093/jmcb/mjab066] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 12/03/2022] Open
Abstract
The cyclic GMP‒AMP synthase (cGAS)‒stimulator of interferon genes (STING) signaling pathway senses the presence of cytosolic DNA and, in turn, triggers downstream signaling to induce the expression of inflammatory and type I interferon genes in immune cells. Whereas the innate immune function of the cGAS‒STING pathway is well studied over the past years, emerging evidence suggests that this signaling pathway may have additional functions beyond innate immune surveillance. Consistent with this notion, dysregulation of the cGAS‒STING signaling pathway in adipocytes, hepatocytes, and renal proximal tubule epithelial cells are associated with metabolic dysfunction, impaired energy homeostasis, and kidney diseases. In this review, we summarize current understanding of the cGAS‒STING pathway in several metabolic diseases such as obesity, insulin resistance, alcoholic and nonalcoholic fatty liver diseases, as well as acute kidney injury and chronic kidney disease. We also review the interaction between the cGAS‒STING pathway and lipid metabolism. Lastly, we discuss potential mechanisms by which cGAS‒STING signaling regulates metabolism and point toward future avenues of research targeting the cGAS‒STING pathway as possible means to treat common metabolic disorders.
Collapse
Affiliation(s)
- Juli Bai
- Departments of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX 78229, USA.,National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Feng Liu
- Departments of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX 78229, USA.,National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
116
|
Kgatle MM, Lawal IO, Mashabela G, Boshomane TMG, Koatale PC, Mahasha PW, Ndlovu H, Vorster M, Rodrigues HG, Zeevaart JR, Gordon S, Moura-Alves P, Sathekge MM. COVID-19 Is a Multi-Organ Aggressor: Epigenetic and Clinical Marks. Front Immunol 2021; 12:752380. [PMID: 34691068 PMCID: PMC8531724 DOI: 10.3389/fimmu.2021.752380] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/21/2021] [Indexed: 12/19/2022] Open
Abstract
The progression of coronavirus disease 2019 (COVID-19), resulting from a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, may be influenced by both genetic and environmental factors. Several viruses hijack the host genome machinery for their own advantage and survival, and similar phenomena might occur upon SARS-CoV-2 infection. Severe cases of COVID-19 may be driven by metabolic and epigenetic driven mechanisms, including DNA methylation and histone/chromatin alterations. These epigenetic phenomena may respond to enhanced viral replication and mediate persistent long-term infection and clinical phenotypes associated with severe COVID-19 cases and fatalities. Understanding the epigenetic events involved, and their clinical significance, may provide novel insights valuable for the therapeutic control and management of the COVID-19 pandemic. This review highlights different epigenetic marks potentially associated with COVID-19 development, clinical manifestation, and progression.
Collapse
Affiliation(s)
- Mankgopo Magdeline Kgatle
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Ismaheel Opeyemi Lawal
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Gabriel Mashabela
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Tebatso Moshoeu Gillian Boshomane
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear and Oncology Division, AXIM Medical (Pty), Midrand
| | - Palesa Caroline Koatale
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Phetole Walter Mahasha
- Precision Medicine and SAMRC Genomic Centre, Grants, Innovation, and Product Development (GIPD) Unit, South African Medical Research Council, Pretoria, South Africa
| | - Honest Ndlovu
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mariza Vorster
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Hosana Gomes Rodrigues
- Laboratory of Nutrients and Tissue Repair, School of Applied Sciences, University of Campinas, Campinas, Brazil
| | - Jan Rijn Zeevaart
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- South African Nuclear Energy Corporation, Radiochemistry and NuMeRI PreClinical Imaging Facility, Mahikeng, South Africa
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Pedro Moura-Alves
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mike Machaba Sathekge
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
117
|
Chen R, Du J, Zhu H, Ling Q. The role of cGAS-STING signalling in liver diseases. JHEP Rep 2021; 3:100324. [PMID: 34381984 PMCID: PMC8340306 DOI: 10.1016/j.jhepr.2021.100324] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/20/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
The recently identified novel cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) activates the downstream adaptor protein stimulator of interferon genes (STING) by catalysing the synthesis of cyclic GMP-AMP. This in turn initiates an innate immune response through the release of various cytokines, including type I interferon. Foreign DNA (microbial infection) or endogenous DNA (nuclear or mitochondrial leakage) can serve as cGAS ligands and lead to the activation of cGAS-STING signalling. Therefore, the cGAS-STING pathway plays essential roles in infectious diseases, sterile inflammation, tumours, and autoimmune diseases. In addition, cGAS-STING signalling affects the progression of liver inflammation through other mechanisms, such as autophagy and metabolism. In this review, we summarise recent advances in our understanding of the role of cGAS-STING signalling in the innate immune modulation of different liver diseases. Furthermore, we discuss the therapeutic potential of targeting the cGAS-STING pathway in the treatment of liver diseases.
Collapse
Key Words
- AIM2, absent in melanoma 2
- ALD, alcohol-related liver disease
- APCs, antigen-presenting cells
- CDNs, cyclic dinucleotides
- DAMPs, damage-associated molecular patterns
- DCs, dendritic cells
- ER, endoplasmic reticulum
- GVHD, graft-versus-host disease
- HCC, hepatocellular carcinoma
- HSCs, hepatic stellate cells
- IFN-I, type I interferon
- IL, interleukin
- IRF3, interferon regulatory factor 3
- IRI, ischaemia refusion injury
- KCs, Kupffer cells
- LSECs, liver sinusoidal endothelial cells
- MHC, major histocompatibility complex
- NAFLD, non-alcoholic fatty liver disease
- NK cells, natural killer cells
- NPCs, non-parenchymal cells
- PAMPs, pathogen-associated molecular patterns
- PD-1, programmed cell death protein-1
- PD-L1, programmed cell death protein ligand-1
- PPRs, pattern recognition receptors
- SAVI, STING-associated vasculopathy with onset in infancy
- STING, stimulator of interferon genes
- TBK1, TANK-binding kinase 1
- TGF-β1, transforming growth factor-β1
- TLR, Toll-like receptor
- TNF, tumour necrosis factor
- XRCC, X-ray repair cross complementing
- aHSCT, allogeneic haematopoietic stem cell transplantation
- cGAMP, cyclic guanosine monophosphate-adenosine monophosphate
- cGAS, cyclic guanosine monophosphate-adenosine monophosphate synthase
- cGAS-STING signalling
- dsDNA, double-strand DNA
- hepatocellular carcinoma
- innate immune response
- liver injury
- mTOR, mammalian target of rapamycin
- mtDNA, mitochondrial DNA
- nonalcoholic fatty liver disease
- siRNA, small interfering RNA
- ssRNA, single-stranded RNA
- viral hepatitis
Collapse
Affiliation(s)
- Ruihan Chen
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiamin Du
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qi Ling
- Department of Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
118
|
Wang L, Zhang S, Han J, Nie X, Qi Y, Han Y, Chen X, He C. Activation of STING Pathway Contributed to Cisplatin-Induced Cardiac Dysfunction via Promoting the Activation of TNF-α-AP-1 Signal Pathway. Front Pharmacol 2021; 12:711238. [PMID: 34483919 PMCID: PMC8415915 DOI: 10.3389/fphar.2021.711238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/03/2021] [Indexed: 01/31/2023] Open
Abstract
Cardiovascular complications are a well-documented limitation of conventional cancer chemotherapy. As a notable side effect of cisplatin, cardiotoxicity represents a major obstacle to the treatment of cancer. Recently, it has been reported that cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) signaling pathway was associated with the occurrence and development of cardiovascular diseases. However, the effect of STING on cardiac damage caused by cisplatin remains unclear. In this study, cisplatin was shown to activate the cGAS-STING signaling pathway, and deficiency of STING attenuated cisplatin-induced cardiotoxicity in vivo and in vitro. Mechanistically, the STING-TNF-α-AP-1 axis contributed to cisplatin-induced cardiotoxicity by triggering cardiomyocyte apoptosis. In conclusion, our results indicated that STING might be a critical regulator of cisplatin-induced cardiotoxicity and be considered as a potential therapeutic target for preventing the progression of chemotherapy-associated cardiovascular complications.
Collapse
Affiliation(s)
- Lintao Wang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Suya Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jibo Han
- Department of Cardiology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaoyan Nie
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yajun Qi
- Department of Pharmacy, Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Yingying Han
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiong Chen
- Department of Endocrinology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chaoyong He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
119
|
Schuliga M, Read J, Knight DA. Ageing mechanisms that contribute to tissue remodeling in lung disease. Ageing Res Rev 2021; 70:101405. [PMID: 34242806 DOI: 10.1016/j.arr.2021.101405] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Age is a major risk factor for chronic respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and certain phenotypes of asthma. The recent COVID-19 pandemic also highlights the increased susceptibility of the elderly to acute respiratory distress syndrome (ARDS), a diffuse inflammatory lung injury with often long-term effects (ie parenchymal fibrosis). Collectively, these lung conditions are characterized by a pathogenic reparative process that, rather than restoring organ function, contributes to structural and functional tissue decline. In the ageing lung, the homeostatic control of wound healing following challenge or injury has an increased likelihood of being perturbed, increasing susceptibility to disease. This loss of fidelity is a consequence of a diverse range of underlying ageing mechanisms including senescence, mitochondrial dysfunction, proteostatic stress and diminished autophagy that occur within the lung, as well as in other tissues, organs and systems of the body. These ageing pathways are highly interconnected, involving localized and systemic increases in inflammatory mediators and damage associated molecular patterns (DAMPs); along with corresponding changes in immune cell function, metabolism and composition of the pulmonary and gut microbiomes. Here we comprehensively review the roles of ageing mechanisms in the tissue remodeling of lung disease.
Collapse
Affiliation(s)
- Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| | - Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
120
|
Akhmetova K, Balasov M, Chesnokov I. Drosophila STING protein has a role in lipid metabolism. eLife 2021; 10:e67358. [PMID: 34467853 PMCID: PMC8443252 DOI: 10.7554/elife.67358] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 08/31/2021] [Indexed: 12/27/2022] Open
Abstract
Stimulator of interferon genes (STING) plays an important role in innate immunity by controlling type I interferon response against invaded pathogens. In this work, we describe a previously unknown role of STING in lipid metabolism in Drosophila. Flies with STING deletion are sensitive to starvation and oxidative stress, have reduced lipid storage, and downregulated expression of lipid metabolism genes. We found that Drosophila STING interacts with lipid synthesizing enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN). ACC and FASN also interact with each other, indicating that all three proteins may be components of a large multi-enzyme complex. The deletion of Drosophila STING leads to disturbed ACC localization and decreased FASN enzyme activity. Together, our results demonstrate a previously undescribed role of STING in lipid metabolism in Drosophila.
Collapse
Affiliation(s)
- Katarina Akhmetova
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, School of MedicineBirminghamUnited States
| | - Maxim Balasov
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, School of MedicineBirminghamUnited States
| | - Igor Chesnokov
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, School of MedicineBirminghamUnited States
| |
Collapse
|
121
|
Shen P, Han L, Chen G, Cheng Z, Liu Q. Emodin Attenuates Acetaminophen-Induced Hepatotoxicity via the cGAS-STING Pathway. Inflammation 2021; 45:74-87. [PMID: 34409550 DOI: 10.1007/s10753-021-01529-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/27/2021] [Indexed: 01/24/2023]
Abstract
Emodin is a natural bioactive compound from traditional Chinese herbs that exerts anti-inflammatory, antioxidant, anticancer, hepatoprotective, and neuroprotective effects. However, the protective effects of emodin in acetaminophen (APAP)-induced hepatotoxicity are not clear. The present study examined the effects of emodin on APAP-induced hepatotoxicity and investigated the potential molecular mechanisms. C57BL/6 mice were pretreated with emodin (15 and 30 mg/kg) for 5 consecutive days and then given APAP (300 mg/kg) to establish an APAP-induced liver injury model. Mice were sacrificed to detect the serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and albumin (ALB) and the liver tissue levels of glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD). Histological assessment, Western blotting, and ELISA were performed. Emodin pretreatment significantly reduced the levels of ALT, AST, and ALP; increased the levels of ALB; alleviated hepatocellular damage and apoptosis; attenuated the exhaustion of GSH and SOD and the accumulation of MDA; and increased the expression of antioxidative enzymes, including nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase 1 (HO-1), and NAD(P)H quinone dehydrogenase 1 (NQO1). Emodin also inhibited the expression of NLRP3 and reduced the levels of pro-inflammatory factors, including interleukin-1 beta (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α). Emodin inhibited interferon (IFN)-α, cyclic GMP-AMP synthase (cGAS), and its downstream signaling effector stimulator of interferon genes (STING) expression to protect the liver against APAP-induced inflammatory responses and apoptosis. These results suggest that emodin protected hepatocytes from APAP-induced liver injury via the upregulation of the Nrf2-mediated antioxidative stress pathway, the inhibition of the NLRP3 inflammasome, and the downregulation of the cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Pan Shen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Liang Han
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Guang Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Zhe Cheng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Qiong Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China.
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
122
|
Xing Y, Xu C, Lin X, Zhao M, Gong D, Liu L, Geng T. Complement C3 participates in the development of goose fatty liver potentially by regulating the expression of FASN and ETNK1. Anim Sci J 2021; 92:e13527. [PMID: 33615637 DOI: 10.1111/asj.13527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/24/2020] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) occurs in humans, domestic animals and poultry. Different from upregulation of complement C3 in human NAFLD, C3 expression is inhibited in goose fatty liver (GFL), implying a specific role of C3 in GFL. This study was mainly focused on uncovering the uniqueness of goose liver cells in the regulation of C3 expression and identifying the downstream genes of C3 to improve understanding on the specific role of C3 in GFL. The results showed that C3 expression was inhibited in the liver, muscle and fat tissues of the overfed versus control (normally fed) geese. Oleate and insulin could inhibit C3 expression in goose primary hepatocytes but induce it in mouse primary hepatocytes. A total of 1,123 differentially expressed genes (DEGs) were affected by C3 overexpression and were mainly enriched in immune response/inflammation and catabolism-related KEGG pathways. Additionally, the representative downstream genes (FASN and ETNK1) of C3 could mediate the role of C3 in the development of GFL. In conclusion, the suppression of C3 in GFL is at least partially attributed to hyperinsulinemia, hyperlipidemia and uniqueness of goose liver cells. Complement C3 does not only affect hepatic steatosis but also affect inflammation/immune response in GFL.
Collapse
Affiliation(s)
- Ya Xing
- Institute of Epigenetics and Epigenomics, Yangzhou University, Yangzhou, P. R. China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Cheng Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Xiao Lin
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Minmeng Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Long Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Tuoyu Geng
- Institute of Epigenetics and Epigenomics, Yangzhou University, Yangzhou, P. R. China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| |
Collapse
|
123
|
Abstract
The cyclic GMP-AMP (cGAMP) synthase (cGAS) has been identified as a cytosolic double stranded DNA sensor that plays a pivotal role in the type I interferon and inflammation responses via the STING-dependent signaling pathway. In the past several years, a growing body of evidence has revealed that cGAS is also localized in the nucleus where it is associated with distinct nuclear substructures such as nucleosomes, DNA replication forks, the double-stranded breaks, and centromeres, suggesting that cGAS may have other functions in addition to its role in DNA sensing. However, while the innate immune function of cGAS is well established, the non-canonical nuclear function of cGAS remains poorly understood. Here, we review our current understanding of the complex nature of nuclear cGAS and point to open questions on the novel roles and the mechanisms of action of this protein as a key regulator of cell nuclear function, beyond its well-established role in dsDNA sensing and innate immune response.
Collapse
Affiliation(s)
- Juli Bai
- Departments of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, USA. .,National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Feng Liu
- Departments of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, USA. .,National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
124
|
Yang GH, Fontaine DA, Lodh S, Blumer JT, Roopra A, Davis DB. TCF19 Impacts a Network of Inflammatory and DNA Damage Response Genes in the Pancreatic β-Cell. Metabolites 2021; 11:metabo11080513. [PMID: 34436454 PMCID: PMC8400192 DOI: 10.3390/metabo11080513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Transcription factor 19 (TCF19) is a gene associated with type 1 diabetes (T1DM) and type 2 diabetes (T2DM) in genome-wide association studies. Prior studies have demonstrated that Tcf19 knockdown impairs β-cell proliferation and increases apoptosis. However, little is known about its role in diabetes pathogenesis or the effects of TCF19 gain-of-function. The aim of this study was to examine the impact of TCF19 overexpression in INS-1 β-cells and human islets on proliferation and gene expression. With TCF19 overexpression, there was an increase in nucleotide incorporation without any change in cell cycle gene expression, alluding to an alternate process of nucleotide incorporation. Analysis of RNA-seq of TCF19 overexpressing cells revealed increased expression of several DNA damage response (DDR) genes, as well as a tightly linked set of genes involved in viral responses, immune system processes, and inflammation. This connectivity between DNA damage and inflammatory gene expression has not been well studied in the β-cell and suggests a novel role for TCF19 in regulating these pathways. Future studies determining how TCF19 may modulate these pathways can provide potential targets for improving β-cell survival.
Collapse
Affiliation(s)
- Grace H. Yang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Danielle A. Fontaine
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Sukanya Lodh
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - Joseph T. Blumer
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Dawn Belt Davis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
125
|
Ning L, Rui X, Bo W, Qing G. The critical roles of histone deacetylase 3 in the pathogenesis of solid organ injury. Cell Death Dis 2021; 12:734. [PMID: 34301918 PMCID: PMC8302660 DOI: 10.1038/s41419-021-04019-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Histone deacetylase 3 (HDAC3) plays a crucial role in chromatin remodeling, which, in turn, regulates gene transcription. Hence, HDAC3 has been implicated in various diseases, including ischemic injury, fibrosis, neurodegeneration, infections, and inflammatory conditions. In addition, HDAC3 plays vital roles under physiological conditions by regulating circadian rhythms, metabolism, and development. In this review, we summarize the current knowledge of the physiological functions of HDAC3 and its role in organ injury. We also discuss the therapeutic value of HDAC3 in various diseases.
Collapse
Affiliation(s)
- Li Ning
- grid.412632.00000 0004 1758 2270Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xiong Rui
- grid.412632.00000 0004 1758 2270Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Wang Bo
- grid.412632.00000 0004 1758 2270Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Geng Qing
- grid.412632.00000 0004 1758 2270Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| |
Collapse
|
126
|
Liu J, Jia Z, Gong W. Circulating Mitochondrial DNA Stimulates Innate Immune Signaling Pathways to Mediate Acute Kidney Injury. Front Immunol 2021; 12:680648. [PMID: 34248963 PMCID: PMC8264283 DOI: 10.3389/fimmu.2021.680648] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/07/2021] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial dysfunction is increasingly considered as a critical contributor to the occurrence and progression of acute kidney injury (AKI). However, the mechanisms by which damaged mitochondria mediate AKI progression are multifactorial and complicated. Mitochondrial DNA (mtDNA) released from damaged mitochondria could serve as a danger-associated molecular pattern (DAMP) and activate the innate immune system through STING, TLR9, NLRP3, and some other adaptors, and further mediate tubular cell inflammation and apoptosis. Accumulating evidence has demonstrated the important role of circulating mtDNA and its related pathways in the progression of AKI, and regulating the proteins involved in these pathways may be an effective strategy to reduce renal tubular injury and alleviate AKI. Here, we aim to provide a comprehensive overview of recent studies on mtDNA-mediated renal pathological events to provide new insights in the setting of AKI.
Collapse
Affiliation(s)
- Jiaye Liu
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Gong
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
127
|
Chen L, Zhao S, Zhu Y, Liu Y, Li H, Zhao Q. Molecular Dynamics Simulations Reveal the Modulated Mechanism of STING Conformation. Interdiscip Sci 2021; 13:751-765. [PMID: 34142362 DOI: 10.1007/s12539-021-00446-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 11/29/2022]
Abstract
Stimulator of interferon genes (STING), which is an integral ER-membrane protein, could induce an antiviral state and boost antitumor immunity. Recent experiments reported that different small molecules could modulate the conformation of the STING. However, the mechanism of small molecules modulating the conformation of STING is still unknown. To illustrate the conformational modulated mechanism of STING by small molecules at atomic level, we investigated the interactions between STING and the small molecules: cGAMP and diABZI with molecular dynamics (MD) simulations method. Interestingly, we found that the residues of STING in the binding pocket are more flexible in the monomers of STING than that in the dimer of STING. We also demonstrated that cGAMP and diABZI have a similar binding mode to STING monomers/dimer, and π-π stacking interactions play important roles for the agonists and STING. Our study proposed mechanistic insights into the STING conformation modulated by small molecules and we suggested that the special molecule (e. g. diABZI) could induce the conformational transition of STING from the "open" monomers to the "closed" dimer state. Our research may provide a clue for the development of cancer immunotherapy.
Collapse
Affiliation(s)
- Li Chen
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 200090, China
| | - Shuang Zhao
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 200090, China
| | - Yanyan Zhu
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 200090, China
| | - Yongsheng Liu
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 200090, China
| | - Huiyu Li
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 200090, China.
| | - Qingjie Zhao
- Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
128
|
Lin Y, Wang L, Ye M, Yu KN, Sun X, Xue M, Deng X. Activation of the cGAS-STING signaling pathway in adenomyosis patients. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:932-942. [PMID: 34010983 PMCID: PMC8342233 DOI: 10.1002/iid3.452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
Objective Adenomyosis is characterized by the presence of endometrium or endometrium‐like glands and stroma within the myometrium. In this study, we aimed to investigate whether the cGAS–STING pathway was activated and correlated with clinical outcomes in adenomyosis patients. Materials and Methods Twenty patients diagnosed with adenomyosis and 10 patients diagnosed with cervical intraepithelial neoplasia grade 3 (CIN‐3) but no adenomyosis were enrolled in this study. Specimens were collected during surgery from August 2017 to December 2017 at Third Xiangya Hospital. The messenger RNA (mRNA) and protein levels of key cGAS–STING pathway factors in uterine tissue were detected by real‐time reverse‐transcription polymerase chain reaction and immunohistochemistry, respectively. The correlations of gene expression and clinical outcomes, including dysmenorrhea and uterine volume, were analyzed. Results The cGAS, STING, TANK‐binding kinase 1 (TBK‐1), interferon‐α (IFN‐α), IFN‐β, and tumor necrosis factor‐α (TNF‐α) mRNA and protein levels in the ectopic endometrial tissue from adenomyosis patients were significantly higher compared with that from the controls in endometrium (p < .05). cGAS and STING gene expression were correlated with TBK‐1, IFN‐β, and TNF‐α expression (p < .05). Importantly, TBK‐1 and TNF‐α expression were correlated with the clinical outcome of dysmenorrhea (p < .05). Conclusion Our study reveals that the cGAS–STING pathway is activated in adenomyosis patients and its activation is subsequently correlated with clinical outcomes, which suggests that the cGAS–STING pathway may contribute to adenomyosis pathogenesis.
Collapse
Affiliation(s)
- Yun Lin
- Department of Obstetrics and Gynecology, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Luying Wang
- Department of Obstetrics and Gynecology, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mingzhu Ye
- Department of Obstetrics and Gynecology, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ke-Nan Yu
- Department of Obstetrics and Gynecology, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xin Sun
- Department of Obstetrics and Gynecology, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Min Xue
- Department of Obstetrics and Gynecology, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xinliang Deng
- Department of Obstetrics and Gynecology, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
129
|
Yu L, Liu P. Cytosolic DNA sensing by cGAS: regulation, function, and human diseases. Signal Transduct Target Ther 2021; 6:170. [PMID: 33927185 PMCID: PMC8085147 DOI: 10.1038/s41392-021-00554-y] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/17/2021] [Accepted: 03/08/2021] [Indexed: 12/16/2022] Open
Abstract
Sensing invasive cytosolic DNA is an integral component of innate immunity. cGAS was identified in 2013 as the major cytosolic DNA sensor that binds dsDNA to catalyze the synthesis of a special asymmetric cyclic-dinucleotide, 2'3'-cGAMP, as the secondary messenger to bind and activate STING for subsequent production of type I interferons and other immune-modulatory genes. Hyperactivation of cGAS signaling contributes to autoimmune diseases but serves as an adjuvant for anticancer immune therapy. On the other hand, inactivation of cGAS signaling causes deficiency to sense and clear the viral and bacterial infection and creates a tumor-prone immune microenvironment to facilitate tumor evasion of immune surveillance. Thus, cGAS activation is tightly controlled. In this review, we summarize up-to-date multilayers of regulatory mechanisms governing cGAS activation, including cGAS pre- and post-translational regulations, cGAS-binding proteins, and additional cGAS regulators such as ions and small molecules. We will also reveal the pathophysiological function of cGAS and its product cGAMP in human diseases. We hope to provide an up-to-date review for recent research advances of cGAS biology and cGAS-targeted therapies for human diseases.
Collapse
Affiliation(s)
- Le Yu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
130
|
Zamorano Cuervo N, Fortin A, Caron E, Chartier S, Grandvaux N. Pinpointing cysteine oxidation sites by high-resolution proteomics reveals a mechanism of redox-dependent inhibition of human STING. Sci Signal 2021; 14:14/680/eaaw4673. [PMID: 33906974 DOI: 10.1126/scisignal.aaw4673] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein function is regulated by posttranslational modifications (PTMs), among which reversible oxidation of cysteine residues has emerged as a key regulatory mechanism of cellular responses. Given the redox regulation of virus-host interactions, the identification of oxidized cysteine sites in cells is essential to understand the underlying mechanisms involved. Here, we present a proteome-wide identification of reversibly oxidized cysteine sites in oxidant-treated cells using a maleimide-based bioswitch method coupled to mass spectrometry analysis. We identified 2720 unique oxidized cysteine sites within 1473 proteins with distinct abundances, locations, and functions. Oxidized cysteine sites were found in numerous signaling pathways, many relevant to virus-host interactions. We focused on the oxidation of STING, the central adaptor of the innate immune type I interferon pathway, which is stimulated in response to the detection of cytosolic DNA by cGAS. We demonstrated the reversible oxidation of Cys148 and Cys206 of STING in cells. Molecular analyses led us to establish a model in which Cys148 oxidation is constitutive, whereas Cys206 oxidation is inducible by oxidative stress or by the natural ligand of STING, 2'3'-cGAMP. Our data suggest that the oxidation of Cys206 prevented hyperactivation of STING by causing a conformational change associated with the formation of inactive polymers containing intermolecular disulfide bonds. This finding should aid the design of therapies targeting STING that are relevant to autoinflammatory disorders, immunotherapies, and vaccines.
Collapse
Affiliation(s)
- Natalia Zamorano Cuervo
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Audray Fortin
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Elise Caron
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Stéfany Chartier
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Nathalie Grandvaux
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada. .,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, H3C 3J7 Québec, Canada
| |
Collapse
|
131
|
Negi CK, Khan S, Dirven H, Bajard L, Bláha L. Flame Retardants-Mediated Interferon Signaling in the Pathogenesis of Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms22084282. [PMID: 33924165 PMCID: PMC8074384 DOI: 10.3390/ijms22084282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing concern worldwide, affecting 25% of the global population. NAFLD is a multifactorial disease with a broad spectrum of pathology includes steatosis, which gradually progresses to a more severe condition such as nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and eventually leads to hepatic cancer. Several risk factors, including exposure to environmental toxicants, are involved in the development and progression of NAFLD. Environmental factors may promote the development and progression of NAFLD by various biological alterations, including mitochondrial dysfunction, reactive oxygen species production, nuclear receptors dysregulation, and interference in inflammatory and immune-mediated signaling. Moreover, environmental contaminants can influence immune responses by impairing the immune system’s components and, ultimately, disease susceptibility. Flame retardants (FRs) are anthropogenic chemicals or mixtures that are being used to inhibit or delay the spread of fire. FRs have been employed in several household and outdoor products; therefore, human exposure is unavoidable. In this review, we summarized the potential mechanisms of FRs-associated immune and inflammatory signaling and their possible contribution to the development and progression of NAFLD, with an emphasis on FRs-mediated interferon signaling. Knowledge gaps are identified, and emerging pharmacotherapeutic molecules targeting the immune and inflammatory signaling for NAFLD are also discussed.
Collapse
Affiliation(s)
- Chander K. Negi
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
- Correspondence: or
| | - Sabbir Khan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA;
| | - Hubert Dirven
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, 0456 Oslo, Norway;
| | - Lola Bajard
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
| | - Luděk Bláha
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
| |
Collapse
|
132
|
Myakala K, Jones BA, Wang XX, Levi M. Sacubitril/valsartan treatment has differential effects in modulating diabetic kidney disease in db/db mice and KKAy mice compared with valsartan treatment. Am J Physiol Renal Physiol 2021; 320:F1133-F1151. [PMID: 33870733 DOI: 10.1152/ajprenal.00614.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although renin-angiotensin blockade has shown beneficial outcomes in patients with diabetes, renal injury progresses in most of these patients. Therefore, there remains a need for new therapeutic targets in diabetic kidney disease. Enhancement of vasoactive peptides, such as natriuretic peptides, via neprilysin inhibition, has been a new approach. A first-in-class drug, sacubitril/valsartan (Sac/Val), a combination of the angiotensin II receptor blocker Val and neprilysin inhibitor prodrug Sac, has been shown to be more effective than renin-angiotensin blockade alone in the treatment of heart failure with reduced ejection fraction. In this study, we tested the effects of Sac/Val in diabetic kidney disease. We administered Sac/Val or Val to two type 2 diabetes mouse models, db/db mice or KKAy mice. After 3 mo of treatment, Sac/Val attenuated the progression of proteinuria, glomerulosclerosis, and podocyte loss in both models of diabetic mice. Val shared a similar improvement but to a lesser degree in some parameters compared with Sac/Val. Sac/Val but not Val decreased the blood glucose level in KKAy mice. Sac/Val exerted renal protection through coordinated effects on antioxidative stress and anti-inflammation. In both diabetic models, we revealed a new mechanism to cause inflammation, self-DNA-activated cGMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling, which was activated in diabetic kidneys and prevented by Sac/Val or Val treatment. The present data suggest that Sac/Val has sufficient therapeutical potential to counter the pathophysiological effects of diabetic kidney disease, and its effectiveness could be better than Val alone.NEW & NOTEWORTHY The first-in-class drug sacubitril/valsartan, a combination of the angiotensin II receptor blocker valsartan and neprilysin inhibitor sacubitril, was tested for its effects in diabetic kidney disease using db/db mice and KKAy mice. We found that Sac/Val has sufficient therapeutical potential to counter the pathophysiological effects of diabetic kidney disease. We further revealed a new mechanism to cause inflammation, self-DNA-activated cGAS-STING signaling, which was activated in diabetic kidneys and prevented by sacubitril/valsartan or valsartan treatment.
Collapse
Affiliation(s)
- Komuraiah Myakala
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia
| | - Bryce A Jones
- Department of Pharmacology and Physiology, Georgetown University, Washington, District of Columbia
| | - Xiaoxin X Wang
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia
| |
Collapse
|
133
|
Chen C, Tong Y, Zheng Y, Shi Y, Chen Z, Li J, Liu X, Zhang D, Yang H. Cytosolic Delivery of Thiolated Mn-cGAMP Nanovaccine to Enhance the Antitumor Immune Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006970. [PMID: 33719177 DOI: 10.1002/smll.202006970] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/07/2021] [Indexed: 06/12/2023]
Abstract
As a stimulator of interferon gene (STING), cyclic dinucleotide activates a broad cellular immune response for anti-cancer immunotherapy (CIT). However, the inherent of instability of 2' 3'-cyclic-GMP-AMP (cGAMP) with poor cellular targeting, rapid clearance, and inefficient transport to the cytoplasm seriously hinders cGAMP potency. Here, a thiolated and Mn2+ coordinated cyclic dinucleotide nanovaccine (termed as Mn-cGAMP NVs) to enable direct cytosolic co-delivery of cGAMP and Mn2+ to potentiate the antitumor immune response is presented. In the NVs, the fixation cGAMP with Mn2+ ions not only improve its stability, but also potentiate the activation of STING. Meanwhile, the presence of polysulfides on the NVs surface allowed direct cytosolic delivery while avoiding degradation. In this way, the production of cytokines for activating T cells immunity is greatly elevated, which in turn suppressed the primary and distal tumors growth through long-term immune memory and led to long-term survival of poorly immunogenic B16F10 melanoma mice. Moreover, by further combining with anti-PD-L1 monoclonal antibody, synergistic T cells antitumor immune response is elicited. This work offers a promising strategy to enhance the potency of cGAMP, holding a considerable potential for CIT applications.
Collapse
Affiliation(s)
- Chengyun Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Yuhong Tong
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Yingjun Shi
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Zhaowei Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Da Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| |
Collapse
|
134
|
FGF21 facilitates autophagy in prostate cancer cells by inhibiting the PI3K-Akt-mTOR signaling pathway. Cell Death Dis 2021; 12:303. [PMID: 33753729 PMCID: PMC7985321 DOI: 10.1038/s41419-021-03588-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor 21 (FGF21) plays an important role in regulating glucose and lipid metabolism, but its role in cancer is less well-studied. We aimed to investigate the action of FGF21 in the development of prostate cancer (PCa). Herein, we found that FGF21 expression was markedly downregulated in PCa tissues and cell lines. FGF21 inhibited the proliferation and clone formation of LNCaP cells (a PCa cell line) and promoted apoptosis. FGF21 also inhibited PCa cell migration and invasiveness. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that FGF21 was related to autophagy and the phosphatidylinositol 3-kinase–Akt kinase–mammalian target of rapamycin (PI3K–Akt–mTOR) pathway. Mechanistically, FGF21 promoted autophagy in LNCaP cells by inhibiting the PI3K–Akt–mTOR–70S6K pathway. In addition, FGF21 inhibited PCa tumorigenesis in vivo in nude mice. Altogether, our findings show that FGF21 inhibits PCa cell proliferation and promoted apoptosis in PCa cells through facilitated autophagy. Therefore, FGF21 might be a potential novel target in PCa therapy.
Collapse
|
135
|
Zhou Y, Zhao S, Gao X, Jiang S, Ma J, Wang R, Li Q, Qin L, Tong Z, Wu J, Zhao J. Staphylococcus aureus Induces IFN-β Production via a CARMA3-Independent Mechanism. Pathogens 2021; 10:pathogens10030300. [PMID: 33806598 PMCID: PMC8000617 DOI: 10.3390/pathogens10030300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
Type I interferon (IFN) induction is a critical component of innate immune response to viral and bacterial infection, including S. aureus, but whether it activates the signaling in macrophages and the regulation mechanisms is less well understood. Here we show that S. aureus infection promoted the IFN-β mRNA expression and stimulator of IFN genes (STING)/TANK-binding kinase 1 (TBK1)/interferon regulatory factor 3 (IRF3)-dependent production of IFN-β. Infection with S. aureus induced caspase recruitment domain and membrane-associated guanylate kinase-like domain protein 3 (CARMA3) expression at both the mRNA and protein levels. The heat-killed bacteria failed to trigger IRF3 phosphorylation and upregulation of CARMA3 expression. However, overexpression of CARMA3 did not affect phosphorylation of TBK1 or IRF3 in RAW264.7 cells, J774A.1 macrophages, and mouse embryonic fibroblast (MEF) cells. In conclusion, S. aureus infection induces STING/TBK1/IRF3-mediated IFN-β production in a CARMA3-independent manner.
Collapse
Affiliation(s)
- Yang Zhou
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
- Correspondence: (Y.Z.); (J.Z.)
| | - Shasha Zhao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Xiao Gao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Songhong Jiang
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Jialu Ma
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Rui Wang
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Qing Li
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China;
| | - Leiying Qin
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Zhizi Tong
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Junwei Wu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (S.Z.); (X.G.); (S.J.); (J.M.); (R.W.); (L.Q.); (Z.T.); (J.W.)
| | - Jianjun Zhao
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
- College of Animal Science and Technology, Southwest University, Chongqing 402460, China;
- Correspondence: (Y.Z.); (J.Z.)
| |
Collapse
|
136
|
Ferriere A, Santa P, Garreau A, Bandopadhyay P, Blanco P, Ganguly D, Sisirak V. Self-Nucleic Acid Sensing: A Novel Crucial Pathway Involved in Obesity-Mediated Metaflammation and Metabolic Syndrome. Front Immunol 2021; 11:624256. [PMID: 33574823 PMCID: PMC7870860 DOI: 10.3389/fimmu.2020.624256] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/10/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity and overweight are a global health problem affecting almost one third of the world population. There are multiple complications associated with obesity including metabolic syndrome that commonly lead to development of type II diabetes and non-alcoholic fatty liver disease. The development of metabolic syndrome and severe complications associated with obesity is attributed to the chronic low-grade inflammation that occurs in metabolic tissues such as the liver and the white adipose tissue. In recent years, nucleic acids (mostly DNA), which accumulate systemically in obese individuals, were shown to aberrantly activate innate immune responses and thus to contribute to metabolic tissue inflammation. This minireview will focus on (i) the main sources and forms of nucleic acids that accumulate during obesity, (ii) the sensing pathways required for their detection, and (iii) the key cellular players involved in this process. Fully elucidating the role of nucleic acids in the induction of inflammation induced by obesity would promote the identification of new and long-awaited therapeutic approaches to limit obesity-mediated complications.
Collapse
Affiliation(s)
| | - Pauline Santa
- CNRS-UMR 5164, Immunoconcept, Bordeaux University, Bordeaux, France
| | - Anne Garreau
- CNRS-UMR 5164, Immunoconcept, Bordeaux University, Bordeaux, France
| | - Purbita Bandopadhyay
- IICB-Translational Research Unit of Excellence, Division of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Patrick Blanco
- CNRS-UMR 5164, Immunoconcept, Bordeaux University, Bordeaux, France.,Immunology and Immunogenetic Department, Bordeaux University Hospital, Bordeaux, France
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, Division of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Vanja Sisirak
- CNRS-UMR 5164, Immunoconcept, Bordeaux University, Bordeaux, France
| |
Collapse
|
137
|
Smith JA. STING, the Endoplasmic Reticulum, and Mitochondria: Is Three a Crowd or a Conversation? Front Immunol 2021; 11:611347. [PMID: 33552072 PMCID: PMC7858662 DOI: 10.3389/fimmu.2020.611347] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022] Open
Abstract
The anti-viral pattern recognition receptor STING and its partnering cytosolic DNA sensor cGAS have been increasingly recognized to respond to self DNA in multiple pathologic settings including cancer and autoimmune disease. Endogenous DNA sources that trigger STING include damaged nuclear DNA in micronuclei and mitochondrial DNA (mtDNA). STING resides in the endoplasmic reticulum (ER), and particularly in the ER-mitochondria associated membranes. This unique location renders STING well poised to respond to intracellular organelle stress. Whereas the pathways linking mtDNA and STING have been addressed recently, the mechanisms governing ER stress and STING interaction remain more opaque. The ER and mitochondria share a close anatomic and functional relationship, with mutual production of, and inter-organelle communication via calcium and reactive oxygen species (ROS). This interdependent relationship has potential to both generate the essential ligands for STING activation and to regulate its activity. Herein, we review the interactions between STING and mitochondria, STING and ER, ER and mitochondria (vis-à-vis calcium and ROS), and the evidence for 3-way communication.
Collapse
Affiliation(s)
- Judith A Smith
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
138
|
Lemmer IL, Willemsen N, Hilal N, Bartelt A. A guide to understanding endoplasmic reticulum stress in metabolic disorders. Mol Metab 2021; 47:101169. [PMID: 33484951 PMCID: PMC7887651 DOI: 10.1016/j.molmet.2021.101169] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/08/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The global rise of metabolic disorders, such as obesity, type 2 diabetes, and cardiovascular disease, demands a thorough molecular understanding of the cellular mechanisms that govern health or disease. The endoplasmic reticulum (ER) is a key organelle for cellular function and metabolic adaptation and, therefore disturbed ER function, known as "ER stress," is a key feature of metabolic disorders. SCOPE OF REVIEW As ER stress remains a poorly defined phenomenon, this review provides a general guide to understanding the nature, etiology, and consequences of ER stress in metabolic disorders. We define ER stress by its type of stressor, which is driven by proteotoxicity, lipotoxicity, and/or glucotoxicity. We discuss the implications of ER stress in metabolic disorders by reviewing evidence implicating ER phenotypes and organelle communication, protein quality control, calcium homeostasis, lipid and carbohydrate metabolism, and inflammation as key mechanisms in the development of ER stress and metabolic dysfunction. MAJOR CONCLUSIONS In mammalian biology, ER is a phenotypically and functionally diverse platform for nutrient sensing, which is critical for cell type-specific metabolic control by hepatocytes, adipocytes, muscle cells, and neurons. In these cells, ER stress is a distinct, transient state of functional imbalance, which is usually resolved by the activation of adaptive programs such as the unfolded protein response (UPR), ER-associated protein degradation (ERAD), or autophagy. However, challenges to proteostasis also impact lipid and glucose metabolism and vice versa. In the ER, sensing and adaptive measures are integrated and failure of the ER to adapt leads to aberrant metabolism, organelle dysfunction, insulin resistance, and inflammation. In conclusion, the ER is intricately linked to a wide spectrum of cellular functions and is a critical component in maintaining and restoring metabolic health.
Collapse
Affiliation(s)
- Imke L Lemmer
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Nienke Willemsen
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Nazia Hilal
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Technische Universität München, Biedersteiner Str. 29, 80802 München, Germany; Department of Molecular Metabolism, 665 Huntington Avenue, Harvard T.H. Chan School of Public Health, 02115 Boston, MA, USA.
| |
Collapse
|
139
|
Zhou H, Peng X, Hu J, Wang L, Luo H, Zhang J, Zhang Y, Li G, Ji Y, Zhang J, Bai J, Liu M, Zhou Z, Liu F. DsbA-L deficiency in T cells promotes diet-induced thermogenesis through suppressing IFN-γ production. Nat Commun 2021; 12:326. [PMID: 33436607 PMCID: PMC7804451 DOI: 10.1038/s41467-020-20665-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 12/10/2020] [Indexed: 01/17/2023] Open
Abstract
Adipose tissue-resident T cells have been recognized as a critical regulator of thermogenesis and energy expenditure, yet the underlying mechanisms remain unclear. Here, we show that high-fat diet (HFD) feeding greatly suppresses the expression of disulfide-bond A oxidoreductase-like protein (DsbA-L), a mitochondria-localized chaperone protein, in adipose-resident T cells, which correlates with reduced T cell mitochondrial function. T cell-specific knockout of DsbA-L enhances diet-induced thermogenesis in brown adipose tissue (BAT) and protects mice from HFD-induced obesity, hepatosteatosis, and insulin resistance. Mechanistically, DsbA-L deficiency in T cells reduces IFN-γ production and activates protein kinase A by reducing phosphodiesterase-4D expression, leading to increased BAT thermogenesis. Taken together, our study uncovers a mechanism by which T cells communicate with brown adipocytes to regulate BAT thermogenesis and whole-body energy homeostasis. Our findings highlight a therapeutic potential of targeting T cells for the treatment of over nutrition-induced obesity and its associated metabolic diseases.
Collapse
MESH Headings
- Adipocytes, Brown/drug effects
- Adipocytes, Brown/metabolism
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Animals
- Diet, High-Fat
- Down-Regulation/drug effects
- Energy Metabolism/drug effects
- Feeding Behavior
- Glutathione Transferase/deficiency
- Glutathione Transferase/metabolism
- Insulin Resistance
- Interferon-gamma/administration & dosage
- Interferon-gamma/biosynthesis
- Interferon-gamma/pharmacology
- Male
- Mice, Knockout
- Mitochondria/drug effects
- Mitochondria/metabolism
- Obesity/genetics
- Obesity/pathology
- T-Lymphocytes/drug effects
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
- Thermogenesis/drug effects
- Thermogenesis/genetics
- Uncoupling Protein 1/metabolism
- Mice
Collapse
Affiliation(s)
- Haiyan Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Xinyi Peng
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Jie Hu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Liwen Wang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Junyan Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Yacheng Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Guobao Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Yujiao Ji
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Juli Bai
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Feng Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
140
|
Proshkina EN, Solovev IA, Shaposhnikov MV, Moskalev AA. Key Molecular Mechanisms of Aging, Biomarkers, and Potential Interventions. Mol Biol 2021. [DOI: 10.1134/s0026893320060096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
141
|
Qi Y, Ying Y, Zou J, Fang Q, Yuan X, Cao Y, Cai Y, Fu S. Kaempferol attenuated cisplatin-induced cardiac injury via inhibiting STING/NF-κB-mediated inflammation. Am J Transl Res 2020; 12:8007-8018. [PMID: 33437376 PMCID: PMC7791507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/26/2020] [Indexed: 06/12/2023]
Abstract
Cardiovascular complications have been well documented as the downside to conventional cancer chemotherapy. As a notable side effect of cisplatin, cardiotoxicity represents a major obstacle to the successful treatment of cancer. It has been reported that kaempferol (KPF) possesses cardioprotective and anti-inflammatory qualities. However, the effect of KPF on cardiac damage caused by conventional cancer chemotherapy remains unclear. In this study, we clarified the protective effect of KPF on cisplatin-induced heart injury, and conducted in-depth research on the molecular mechanism underlying this effect. The results showed that KPF protected against cardiac dysfunction and injury induced by cisplatin in vivo. In H9c2 cells, KPF dramatically reduced cispaltin-induced apoptosis and inflammatory response by modulating STING/NF-κB pathway. In conclusion, these results showed that KPF had great potential in attenuating cisplatin-induced cardiac injury. Besides, greater emphasis should be placed in the future on natural active compounds containing KPF with anti-inflammatory effects for the treatment of these diseases.
Collapse
Affiliation(s)
- Yajun Qi
- Department of Pharmacy, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310022, Zhejiang, China
| | - Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, Zhejiang, China
| | - Jie Zou
- Department of Pharmacy, The 903rd Hospital of PLAHangzhou 310000, Zhejiang, China
| | - Qilu Fang
- Department of Pharmacy, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310022, Zhejiang, China
| | - Xiaohong Yuan
- Department of Anesthesiology, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310012, Zhejiang, China
| | - Yingying Cao
- Department of Pharmacy, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310022, Zhejiang, China
| | - Yunfang Cai
- Department of Anesthesiology, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310012, Zhejiang, China
| | - Shuang Fu
- Department of Anesthesiology, The Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of SciencesHangzhou 310012, Zhejiang, China
| |
Collapse
|
142
|
Small molecules targeting the innate immune cGAS‒STING‒TBK1 signaling pathway. Acta Pharm Sin B 2020; 10:2272-2298. [PMID: 33354501 PMCID: PMC7745059 DOI: 10.1016/j.apsb.2020.03.001] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple cancer immunotherapies including chimeric antigen receptor T cell and immune checkpoint inhibitors (ICIs) have been successfully developed to treat various cancers by motivating the adaptive anti-tumor immunity. Particularly, the checkpoint blockade approach has achieved great clinic success as evidenced by several U.S. Food and Drug Administration (FDA)-approved anti-programmed death receptor 1/ligand 1 or anti-cytotoxic T lymphocyte associated protein 4 antibodies. However, the majority of cancers have low clinical response rates to these ICIs due to poor tumor immunogenicity. Indeed, the cyclic guanosine monophosphate-adenosine monophosphate synthase‒stimulator of interferon genes‒TANK-binding kinase 1 (cGAS‒STING‒TBK1) axis is now appreciated as the major signaling pathway in innate immune response across different species. Aberrant signaling of this pathway has been closely linked to multiple diseases, including auto-inflammation, virus infection and cancers. In this perspective, we provide an updated review on the latest progress on the development of small molecule modulators targeting the cGAS‒STING‒TBK1 signaling pathway and their preclinical and clinical use as a new immune stimulatory therapy. Meanwhile, highlights on the clinical candidates, limitations and challenges, as well as future directions in this field are also discussed. Further, small molecule inhibitors targeting this signaling axis and their potential therapeutic use for various indications are discussed as well.
Collapse
Key Words
- ABZI, amidobenzimidazole
- ACMA, 9-amino-6-chloro-2-methoxyacridine
- AMP, adenosine monophosphate
- ATP, adenosine triphosphate
- Anti-tumor
- BNBC, 6-bromo-N-(naphthalen-1-yl)benzo[d][1,3]dioxole-5-carboxamide
- CBD, cyclic dinucleotide-binding domain
- CDA, cyclic diadenosine monophosphate (c-di-AMP)
- CDG, cyclic diguanosine monophosphate (c-di-GMP)
- CDN, cyclic dinucleotide
- CMA, 10-carboxymethyl-9-acridanone
- CTD, C-terminal domain
- CTLA-4, cytotoxic T lymphocyte associated protein 4
- CTT, C-terminal tail
- CXCL, chemokine (C-X-C motif) ligand
- DC50, concentration for 50% degradation
- DCs, dendritic cells
- DMXAA, 5,6-dimethylxanthenone-4-acetic acid
- DSDP, dispiro diketopiperzine
- EM, cryo-electron microscopy
- ENPP1, ecto-nucleotide pyrophosphatase/phosphodiesterase
- ER, endoplasmic reticulum
- FAA, flavone-8-acetic acid
- FDA, U.S. Food and Drug Administration
- FP, fluorescence polarization
- GMP, guanosine monophosphate
- GTP, guanosine triphosphate
- HCQ, hydrochloroquine
- HTS, high throughput screening
- ICI, immune checkpoint inhibitor
- IKK, IκB kinase
- IO, immune-oncology
- IRF3, interferon regulatory factor 3
- ISG, interferon stimulated gene
- ITC, isothermal titration calorimetry
- Immunotherapy
- KD, kinase domain
- LBD, ligand-binding domain
- MDCK, Madin–Darby canine kidney
- MG, Mangostin
- MI, maximum induction
- MLK, mixed lineage kinase
- MinEC5×, minimum effective concentration for inducing 5-fold luciferase activity
- NF-κB, nuclear factor-κB
- Ntase, nucleotidyl transferase
- PBMCs, peripheral-blood mononuclear cells
- PD-1, programmed death receptor 1
- PD-L1, programmed death ligand 1
- PDE, phosphodiesterases
- PDK1, 3-phosphoinositide-dependent protein kinase 1
- PPi, pyrophosphoric acid
- PROTACs, proteolysis targeting chimeras
- PRRs, pattern recognition receptors
- QC, quinacrine
- SAR, structure–activity relationship
- SDD, scaffold and dimerization domain
- STAT, signal transducer and activator of transcription
- STING
- STING, stimulator of interferon genes
- Small molecule modulators
- TBK1
- TBK1, TANK-binding kinase 1
- THIQCs, tetrahydroisoquinolone acetic acids
- TNFRSF, tumor necrosis factor receptor superfamily
- ULD, ubiquitin-like domain
- VHL, von Hippel–Lindau
- cAIMP, cyclic adenosine-inosine monophosphate
- cGAMP, cyclic guanosine monophosphate-adenosine monophosphate
- cGAS
- cGAS, cyclic guanosine monophosphate-adenosine monophosphate synthase
- dsDNA, double-stranded DNA
- i.t., intratumoral
Collapse
|
143
|
Forrester JV, Kuffova L, Delibegovic M. The Role of Inflammation in Diabetic Retinopathy. Front Immunol 2020; 11:583687. [PMID: 33240272 PMCID: PMC7677305 DOI: 10.3389/fimmu.2020.583687] [Citation(s) in RCA: 233] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is central to pathogenic processes in diabetes mellitus and the metabolic syndrome and particularly implicates innate immunity in the development of complications. Inflammation is a primary event in Type 1 diabetes where infectious (viral) and/or autoimmune processes initiate disease; in contrast, chronic inflammation is typical in Type 2 diabetes and is considered a sequel to increasing insulin resistance and disturbed glucose metabolism. Diabetic retinopathy (DR) is perceived as a vascular and neurodegenerative disease which occurs after some years of poorly controlled diabetes. However, many of the clinical features of DR are late events and reflect the nature of the retinal architecture and its cellular composition. Retinal microvascular disease is, in fact, an early event pathogenetically, induced by low grade, persistent leukocyte activation which causes repeated episodes of capillary occlusion and, progressive, attritional retinal ischemia. The later, overt clinical signs of DR are a consequence of the retinal ischemia. Metabolic dysregulation involving both lipid and glucose metabolism may lead to leukocyte activation. On a molecular level, we have shown that macrophage-restricted protein tyrosine phosphatase 1B (PTP1B) is a key regulator of inflammation in the metabolic syndrome involving insulin resistance and it is possible that PTP1B dysregulation may underlie retinal microvascular disease. We have also shown that adherent CCR5+CD11b+ monocyte macrophages appear to be selectively involved in retinal microvascular occlusion. In this review, we discuss the relationship between early leukocyte activation and the later features of DR, common pathogenetic processes between diabetic microvascular disease and other vascular retinopathies, the mechanisms whereby leukocyte activation is induced in hyperglycemia and dyslipidemia, the signaling mechanisms involved in diabetic microvascular disease, and possible interventions which may prevent these retinopathies. We also address a possible role for adaptive immunity in DR. Although significant improvements in treatment of DR have been made with intravitreal anti-VEGF therapy, a sizeable proportion of patients, particularly with sight-threatening macular edema, fail to respond. Alternative therapies targeting inflammatory processes may offer an advantage.
Collapse
Affiliation(s)
- John V Forrester
- Institute of Medical Sciences, University of Aberdeen, Scotland, United Kingdom
| | - Lucia Kuffova
- Institute of Medical Sciences, University of Aberdeen, Scotland, United Kingdom.,Eye Clinic, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Mirela Delibegovic
- Institute of Medical Sciences, University of Aberdeen, Scotland, United Kingdom
| |
Collapse
|
144
|
Mikami A, Erande N, Matsuda S, Kel'in A, Woods LB, Chickering T, Pallan PS, Schlegel MK, Zlatev I, Egli M, Manoharan M. Synthesis, chirality-dependent conformational and biological properties of siRNAs containing 5'-(R)- and 5'-(S)-C-methyl-guanosine. Nucleic Acids Res 2020; 48:10101-10124. [PMID: 32990754 PMCID: PMC7544225 DOI: 10.1093/nar/gkaa750] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 08/10/2020] [Accepted: 08/27/2020] [Indexed: 12/22/2022] Open
Abstract
Various chemical modifications have been identified that enhance potency of small interfering RNAs (siRNAs) and that reduce off-target effects, immune stimulation, and toxicities of metabolites of these therapeutic agents. We previously described 5′-C-methyl pyrimidine nucleotides also modified at the 2′ position of the sugar. Here, we describe the synthesis of 2′-position unmodified 5′-(R)- and 5′-(S)-C-methyl guanosine and evaluation of these nucleotides in the context of siRNA. The (R) isomer provided protection from 5′ exonuclease and the (S) isomer provided protection from 3′ exonuclease in the context of a terminally modified oligonucleotide. siRNA potency was maintained when these modifications were incorporated at the tested positions of sense and antisense strands. Moreover, the corresponding 5′ triphosphates were not substrates for mitochondrial DNA polymerase. Models generated based on crystal structures of 5′ and 3′ exonuclease oligonucleotide complexes with 5′-(R)- and 5′-(S)-C-methyl substituents attached to the 5′- and 3′-terminal nucleotides, respectively, provided insight into the origins of the observed protections. Structural properties of 5′-(R)-C-methyl guanosine incorporated into an RNA octamer were analysed by X-ray crystallography, and the structure explains the loss in duplex thermal stability for the (R) isomer compared with the (S) isomer. Finally, the effect of 5′-C-methylation on endoribonuclease activity has been explained.
Collapse
Affiliation(s)
- Atsushi Mikami
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Namrata Erande
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Shigeo Matsuda
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Alexander Kel'in
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Lauren Blair Woods
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Tyler Chickering
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Pradeep S Pallan
- Department of Biochemistry Vanderbilt University, School of Medicine Nashville, TN 37232, USA
| | - Mark K Schlegel
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Ivan Zlatev
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| | - Martin Egli
- Department of Biochemistry Vanderbilt University, School of Medicine Nashville, TN 37232, USA
| | - Muthiah Manoharan
- Alnylam Pharmaceuticals, 675 West Kendall Street, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
145
|
Amor S, Fernández Blanco L, Baker D. Innate immunity during SARS-CoV-2: evasion strategies and activation trigger hypoxia and vascular damage. Clin Exp Immunol 2020; 202:193-209. [PMID: 32978971 PMCID: PMC7537271 DOI: 10.1111/cei.13523] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 12/18/2022] Open
Abstract
Innate immune sensing of viral molecular patterns is essential for development of antiviral responses. Like many viruses, SARS-CoV-2 has evolved strategies to circumvent innate immune detection, including low cytosine-phosphate-guanosine (CpG) levels in the genome, glycosylation to shield essential elements including the receptor-binding domain, RNA shielding and generation of viral proteins that actively impede anti-viral interferon responses. Together these strategies allow widespread infection and increased viral load. Despite the efforts of immune subversion, SARS-CoV-2 infection activates innate immune pathways inducing a robust type I/III interferon response, production of proinflammatory cytokines and recruitment of neutrophils and myeloid cells. This may induce hyperinflammation or, alternatively, effectively recruit adaptive immune responses that help clear the infection and prevent reinfection. The dysregulation of the renin-angiotensin system due to down-regulation of angiotensin-converting enzyme 2, the receptor for SARS-CoV-2, together with the activation of type I/III interferon response, and inflammasome response converge to promote free radical production and oxidative stress. This exacerbates tissue damage in the respiratory system, but also leads to widespread activation of coagulation pathways leading to thrombosis. Here, we review the current knowledge of the role of the innate immune response following SARS-CoV-2 infection, much of which is based on the knowledge from SARS-CoV and other coronaviruses. Understanding how the virus subverts the initial immune response and how an aberrant innate immune response contributes to the respiratory and vascular damage in COVID-19 may help to explain factors that contribute to the variety of clinical manifestations and outcome of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- S. Amor
- Pathology DepartmentVUMC, Amsterdam UMCAmsterdamthe Netherlands
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University of LondonUK
| | | | - D. Baker
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University of LondonUK
| |
Collapse
|
146
|
Deficiency of β-carotene oxygenase 2 induces mitochondrial fragmentation and activates the STING-IRF3 pathway in the mouse hypothalamus. J Nutr Biochem 2020; 88:108542. [PMID: 33129969 DOI: 10.1016/j.jnutbio.2020.108542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/23/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022]
Abstract
Hypothalamic inflammation has been linked to various aspects of central metabolic dysfunction and diseases in humans, including hyperphagia, altered energy expenditure, and obesity. We previously reported that loss of β-carotene oxygenase 2 (BCO2), a mitochondrial inner membrane protein, causes the alteration of the hypothalamic metabolome, low-grade inflammation, and an increase in food intake in mice at an early age, e.g., 3-6 weeks. Here, we determined the extent to which the deficiency of BCO2 induces hypothalamic inflammation in BCO2 knockout mice. Mitochondrial proteomics, electron microscopy, and immunoblotting were used to assess the changes in hypothalamic mitochondrial dynamics and mitochondrial DNA sensing and signaling. The results showed that deficiency of BCO2 altered hypothalamic mitochondrial proteome and respiratory supercomplex assembly by enhancing the expression of NADH:ubiquinone oxidoreductase subunit A11 protein and improved cardiolipin synthesis. BCO2 deficiency potentiated mitochondrial fission but suppressed mitophagy and mitochondrial biogenesis. Furthermore, deficiency of BCO2 resulted in inactivation of mitochondrial MnSOD enzyme, excessive production of reactive oxygen species, and elevation of protein levels of stimulator of interferon genes (STING) and interferon regulatory factor 3 (IRF3) in the hypothalamus. The data suggest that BCO2 is essential for hypothalamic mitochondrial dynamics. BCO2 deficiency induces mitochondrial fragmentation and mitochondrial oxidative stress, which may lead to mitochondrial DNA release into the cytosol and subsequently sensing by activation of the STING-IRF3 signaling pathway in the mouse hypothalamus.
Collapse
|
147
|
Wang L, Hu J, Zhou H. Macrophage and Adipocyte Mitochondrial Dysfunction in Obesity-Induced Metabolic Diseases. World J Mens Health 2020; 39:606-614. [PMID: 33151047 PMCID: PMC8443980 DOI: 10.5534/wjmh.200163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022] Open
Abstract
Obesity is one of major health burdens of modern society as it contributes to the growing prevalence of its related comorbidities, such as diabetes, cardiovascular diseases, and some cancers. A series of innate immune cells, especially macrophages, and adipocytes have been implicated in the pathogenesis of obesity. Mitochondrial dysfunction, which is induced by obesity, are critical mediators in initiating inflammation in macrophages and adipocytes, and subsequent systemic insulin resistance. In this review, we discuss new findings on how obesity impairs mitochondrial function in macrophages and adipocytes and how this dysfunction contributes to obesity and its comorbidities. We also summarize drugs that treat metabolic diseases by targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Liwen Wang
- Department of Metabolism and Endocrinology, the Second Xiangya Hospital, Central South University, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Hunan, China
| | - Jie Hu
- Department of Metabolism and Endocrinology, the Second Xiangya Hospital, Central South University, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Hunan, China
| | - Haiyan Zhou
- Department of Metabolism and Endocrinology, the Second Xiangya Hospital, Central South University, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Hunan, China.
| |
Collapse
|
148
|
Xu W, Ocak U, Gao L, Tu S, Lenahan CJ, Zhang J, Shao A. Selective autophagy as a therapeutic target for neurological diseases. Cell Mol Life Sci 2020; 78:1369-1392. [PMID: 33067655 PMCID: PMC7904548 DOI: 10.1007/s00018-020-03667-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/03/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
The neurological diseases primarily include acute injuries, chronic neurodegeneration, and others (e.g., infectious diseases of the central nervous system). Autophagy is a housekeeping process responsible for the bulk degradation of misfolded protein aggregates and damaged organelles through the lysosomal machinery. Recent studies have suggested that autophagy, particularly selective autophagy, such as mitophagy, pexophagy, ER-phagy, ribophagy, lipophagy, etc., is closely implicated in neurological diseases. These forms of selective autophagy are controlled by a group of important proteins, including PTEN-induced kinase 1 (PINK1), Parkin, p62, optineurin (OPTN), neighbor of BRCA1 gene 1 (NBR1), and nuclear fragile X mental retardation-interacting protein 1 (NUFIP1). This review highlights the characteristics and underlying mechanisms of different types of selective autophagy, and their implications in various forms of neurological diseases.
Collapse
Affiliation(s)
- Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Umut Ocak
- Department of Emergency Medicine, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, 16310, Bursa, Turkey.,Department of Emergency Medicine, Bursa City Hospital, 16110, Bursa, Turkey
| | - Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | | | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Brain Research Institute, Zhejiang University, Hangzhou, China. .,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China.
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
149
|
Gao M, He Y, Tang H, Chen X, Liu S, Tao Y. cGAS/STING: novel perspectives of the classic pathway. MOLECULAR BIOMEDICINE 2020; 1:7. [PMID: 35006429 PMCID: PMC8603984 DOI: 10.1186/s43556-020-00006-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/09/2020] [Indexed: 02/06/2023] Open
Abstract
Cyclic GMP-AMP (cGAMP) synthase (cGAS) is a cytosolic DNA sensor and innate immune response initiator. Binding with exogenous or endogenous nucleic acids, cGAS activates its downstream adaptor, stimulator of interferon genes (STING). STING then triggers protective immune to enable the elimination of the pathogens and the clearance of cancerous cells. Apparently, aberrantly activated by self-DNA, cGAS/STING pathway is threatening to cause autoimmune and inflammatory diseases. The effects of cGAS/STING in defenses against infection and autoimmune diseases have been well studied, still it is worthwhile to discuss the roles of cGAS/STING pathway beyond the “classical” realm of innate immunity. Recent studies have revealed its involvement in non-canonical inflammasome formation, calcium hemostasis regulation, endoplasmic reticulum (ER) stress response, perception of leaking mitochondrial DNA (mtDNA), autophagy induction, cellular senescence and senescence-associated secretory phenotype (SASP) production, providing an exciting area for future exploration. Previous studies generally focused on the function of cGAS/STING pathway in cytoplasm and immune response. In this review, we summarize the latest research of this pathway on the regulation of other physiological process and STING independent reactions to DNA in micronuclei and nuclei. Together, these studies provide a new perspective of cGAS/STING pathway in human diseases.
Collapse
Affiliation(s)
- Menghui Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yuchen He
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Xiangyu Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China. .,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China. .,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
150
|
The Central Role and Possible Mechanisms of Bacterial DNAs in Sepsis Development. Mediators Inflamm 2020; 2020:7418342. [PMID: 32934605 PMCID: PMC7479481 DOI: 10.1155/2020/7418342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/20/2020] [Indexed: 12/20/2022] Open
Abstract
The pathological roles of bacterial DNA have been documented many decades ago. Bacterial DNAs are different from mammalian DNAs; the latter are heavily methylated. Mammalian cells have sensors such as TLR-9 to sense the DNAs with nonmethylated CpGs and distinguish them from host DNAs with methylated CpGs. Further investigation has identified many other types of DNA sensors distributed in a variety of cellular compartments. These sensors not only sense foreign DNAs, including bacterial and viral DNAs, but also sense damaged DNAs from the host cells. The major downstream signalling pathways includeTLR-9-MyD88-IKKa-IRF-7/NF-κB pathways to increase IFN/proinflammatory cytokine production, STING-TBK1-IRF3 pathway to increase IFN-beta, and AIM2-ASC-caspas-1 pathway to release IL-1beta. The major outcome is to activate host immune response by inducing cytokine production. In this review, we focus on the roles and potential mechanisms of DNA sensors and downstream pathways in sepsis. Although bacterial DNAs play important roles in sepsis development, bacterial DNAs alone are unable to cause severe disease nor lead to death. Priming animals with bacterial DNAs facilitate other pathological factors, such as LPS and other virulent factors, to induce severe disease and lethality. We also discuss compartmental distribution of DNA sensors and pathological significance as well as the transport of extracellular DNAs into cells. Understanding the roles of DNA sensors and signal pathways will pave the way for novel therapeutic strategies in many diseases, particularly in sepsis.
Collapse
|