1
|
Shrestha P, Duwa R, Lee S, Kwon TK, Jeong JH, Yook S. ROS-responsive thioketal nanoparticles delivering system for targeted ulcerative colitis therapy with potent HDAC6 inhibitor, tubastatin A. Eur J Pharm Sci 2024; 201:106856. [PMID: 39032536 DOI: 10.1016/j.ejps.2024.106856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Ulcerative colitis (UC) is a common gastrointestinal problem characterized by the mucosal injury primarily affecting the large intestine. Currently available therapies are not satisfactory as evidenced by high relapse rate and adverse effects. In this study we aimed to develop an effective drug delivery system using reactive oxygen species (ROS)-responsive thioketal nanoparticles (TKNP), to deliver tubastatin A, a potent HDAC6 inhibitor, to the inflamed colon in mice with ulcerative colitis (UC). TKNPs were synthesized by step-growth polymerization from an acetal exchange reaction while TUBA-TKNP was prepared using the single emulsion solvent evaporation technique. Our developed nanoparticle showed release of tubastatin A only in presence of ROS which is found to be highly present at the site of inflamed colon. Oral administration of TUBA-TKNP resulted in the higher accumulation of tubastatin A at the inflamed colon site and decreased the inflammation as evidenced by reduced infiltration of immune cells and decreased level of pro-inflammatory cytokines in TUBA-TKNP treated mice. In summary, our results show the successful localization of tubastatin A at the site of colon inflammation through TUBA-TKNP delivery, as well as resolution of clinical features of UC in mice.
Collapse
Affiliation(s)
- Prabhat Shrestha
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ramesh Duwa
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Radiology, Molecular Imaging Program at Standford (MIPS), School of Medicine, Standford University, Standford, California 94305, USA
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
2
|
Zhang SY, Zhang LY, Wen R, Yang N, Zhang TN. Histone deacetylases and their inhibitors in inflammatory diseases. Biomed Pharmacother 2024; 179:117295. [PMID: 39146765 DOI: 10.1016/j.biopha.2024.117295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Despite considerable research efforts, inflammatory diseases remain a heavy burden on human health, causing significant economic losses annually. Histone deacetylases (HDACs) play a significant role in regulating inflammation (via histone and non-histone protein deacetylation) and chromatin structure and gene expression regulation. Herein, we present a detailed description of the different HDACs and their functions and analyze the role of HDACs in inflammatory diseases, including pro-inflammatory cytokine production reduction, immune cell function modulation, and anti-inflammatory cell activity enhancement. Although HDAC inhibitors have shown broad inflammatory disease treatment potentials, their clinical applicability remains limited because of their non-specific effects, adverse effects, and drug resistance. With further research and insight, these inhibitors are expected to become important tools for the treatment of a wide range of inflammatory diseases. This review aims to explore the mechanisms and application prospects of HDACs and their inhibitors in multiple inflammatory diseases.
Collapse
Affiliation(s)
- Sen-Yu Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Li-Ying Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
3
|
Gurung B, Stricklin M, Wang S. Gut Microbiota-Gut Metabolites and Clostridioides difficile Infection: Approaching Sustainable Solutions for Therapy. Metabolites 2024; 14:74. [PMID: 38276309 PMCID: PMC10819375 DOI: 10.3390/metabo14010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/06/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Clostridioides difficile (C. difficile) infection (CDI) is the most common hospital-acquired infection. With the combination of a high rate of antibiotic resistance and recurrence, it has proven to be a debilitating public health threat. Current treatments for CDI include antibiotics and fecal microbiota transplantation, which contribute to recurrent CDIs and potential risks. Therefore, there is an ongoing need to develop new preventative treatment strategies for CDI. Notably, gut microbiota dysbiosis is the primary risk factor for CDI and provides a promising target for developing novel CDI therapy approaches. Along with gut microbiota dysbiosis, a reduction in important gut metabolites like secondary bile acids and short-chain fatty acids (SCFAs) were also seen in patients suffering from CDI. In this review study, we investigated the roles and mechanisms of gut microbiota and gut microbiota-derived gut metabolites, especially secondary bile acids and SCFAs in CDI pathogenesis. Moreover, specific signatures of gut microbiota and gut metabolites, as well as different factors that can modulate the gut microbiota, were also discussed, indicating that gut microbiota modulators like probiotics and prebiotics can be a potential therapeutic strategy for CDI as they can help restore gut microbiota and produce gut metabolites necessary for a healthy gut. The understanding of the associations between gut microbiota-gut metabolites and CDI will allow for developing precise and sustainable approaches, distinct from antibiotics and fecal transplant, for mitigating CDI and other gut microbiota dysbiosis-related diseases.
Collapse
Affiliation(s)
- Bijay Gurung
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (B.G.); (M.S.)
- Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA
- Interdisciplinary Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
| | - Maranda Stricklin
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (B.G.); (M.S.)
- Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA
| | - Shaohua Wang
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (B.G.); (M.S.)
- Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
4
|
Li M, Peng D, Cao H, Yang X, Li S, Qiu HJ, Li LF. The Host Cytoskeleton Functions as a Pleiotropic Scaffold: Orchestrating Regulation of the Viral Life Cycle and Mediating Host Antiviral Innate Immune Responses. Viruses 2023; 15:1354. [PMID: 37376653 PMCID: PMC10301361 DOI: 10.3390/v15061354] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Viruses are obligate intracellular parasites that critically depend on their hosts to initiate infection, complete replication cycles, and generate new progeny virions. To achieve these goals, viruses have evolved numerous elegant strategies to subvert and utilize different cellular machinery. The cytoskeleton is often one of the first components to be hijacked as it provides a convenient transport system for viruses to enter the cell and reach the site of replication. The cytoskeleton is an intricate network involved in controlling the cell shape, cargo transport, signal transduction, and cell division. The host cytoskeleton has complex interactions with viruses during the viral life cycle, as well as cell-to-cell transmission once the life cycle is completed. Additionally, the host also develops unique, cytoskeleton-mediated antiviral innate immune responses. These processes are also involved in pathological damages, although the comprehensive mechanisms remain elusive. In this review, we briefly summarize the functions of some prominent viruses in inducing or hijacking cytoskeletal structures and the related antiviral responses in order to provide new insights into the crosstalk between the cytoskeleton and viruses, which may contribute to the design of novel antivirals targeting the cytoskeleton.
Collapse
Affiliation(s)
| | | | | | | | | | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| |
Collapse
|
5
|
Hussong SA, Banh AQ, Van Skike CE, Dorigatti AO, Hernandez SF, Hart MJ, Ferran B, Makhlouf H, Gaczynska M, Osmulski PA, McAllen SA, Dineley KT, Ungvari Z, Perez VI, Kayed R, Galvan V. Soluble pathogenic tau enters brain vascular endothelial cells and drives cellular senescence and brain microvascular dysfunction in a mouse model of tauopathy. Nat Commun 2023; 14:2367. [PMID: 37185259 PMCID: PMC10126555 DOI: 10.1038/s41467-023-37840-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Vascular mechanisms of Alzheimer's disease (AD) may constitute a therapeutically addressable biological pathway underlying dementia. We previously demonstrated that soluble pathogenic forms of tau (tau oligomers) accumulate in brain microvasculature of AD and other tauopathies, including prominently in microvascular endothelial cells. Here we show that soluble pathogenic tau accumulates in brain microvascular endothelial cells of P301S(PS19) mice modeling tauopathy and drives AD-like brain microvascular deficits. Microvascular impairments in P301S(PS19) mice were partially negated by selective removal of pathogenic soluble tau aggregates from brain. We found that similar to trans-neuronal transmission of pathogenic forms of tau, soluble tau aggregates are internalized by brain microvascular endothelial cells in a heparin-sensitive manner and induce microtubule destabilization, block endothelial nitric oxide synthase (eNOS) activation, and potently induce endothelial cell senescence that was recapitulated in vivo in microvasculature of P301S(PS19) mice. Our studies suggest that soluble pathogenic tau aggregates mediate AD-like brain microvascular deficits in a mouse model of tauopathy, which may arise from endothelial cell senescence and eNOS dysfunction triggered by internalization of soluble tau aggregates.
Collapse
Affiliation(s)
- Stacy A Hussong
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Oklahoma City Veterans Health Care System, 921 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Andy Q Banh
- South Texas Medical Scientist Training Program, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Candice E Van Skike
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Angela O Dorigatti
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Stephen F Hernandez
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Matthew J Hart
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Therapeutic Science, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Beatriz Ferran
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Haneen Makhlouf
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Maria Gaczynska
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Pawel A Osmulski
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Salome A McAllen
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Kelly T Dineley
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Zoltan Ungvari
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, 800 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, H-1085 Budapest, Üllői út 26, Budapest, Hungary
| | | | - Rakez Kayed
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Veronica Galvan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA.
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Health Care System, 921 NE 13th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
6
|
Xue Y, Gan B, Zhou Y, Wang T, Zhu T, Peng X, Zhang X, Zhou Y. Advances in the Mechanistic Study of the Control of Oxidative Stress Injury by Modulating HDAC6 Activity. Cell Biochem Biophys 2023; 81:127-139. [PMID: 36749475 PMCID: PMC9925596 DOI: 10.1007/s12013-022-01125-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/14/2022] [Indexed: 02/08/2023]
Abstract
Oxidative stress is defined as an injury resulting from a disturbance in the dynamic equilibrium of the redox environment due to the overproduction of active/radical oxygen exceeding the antioxidative ability of the body. This is a key step in the development of various diseases. Oxidative stress is modulated by different factors and events, including the modification of histones, which are the cores of nucleosomes. Histone modification includes acetylation and deacetylation of certain amino acid residues; this process is catalyzed by different enzymes. Histone deacetylase 6 (HDAC6) is a unique deacetylating protease that also catalyzes the deacetylation of different nonhistone substrates to regulate various physiologic processes. The intimate relationship between HDAC6 and oxidative stress has been demonstrated by different studies. The present paper aims to summarize the data obtained from a mechanistic study of HDAC6 and oxidative stress to guide further investigations on mechanistic characterization and drug development.
Collapse
Affiliation(s)
- Yuanye Xue
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China
| | - Bing Gan
- The Third Affiliated Hospital of Guangdong Medical University, Fo Shan, 528000, Guangdong, China
| | - Yanxing Zhou
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Tingyu Wang
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China
| | - Tong Zhu
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China
| | - Xinsheng Peng
- Biomedical Innovation Center, Guangdong Medical University, Dongguan, 523808, China.
- Institute of Marine Medicine, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Xiangning Zhang
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China.
| | - Yanfang Zhou
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
7
|
The role of short-chain fatty acids in Clostridioides difficile infection: A review. Anaerobe 2022; 75:102585. [DOI: 10.1016/j.anaerobe.2022.102585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/20/2022]
|
8
|
Brunetti G, Giuliani A, Navazio AS, Paradisi C, Raponi F, Conti LA, Raponi G. Candida gut colonization, yeast species distribution, and biofilm production in Clostridioides difficile infected patients: a comparison between three populations in two different time periods. Braz J Microbiol 2021; 52:1845-1852. [PMID: 34264501 PMCID: PMC8578342 DOI: 10.1007/s42770-021-00512-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/27/2021] [Indexed: 02/08/2023] Open
Abstract
Candida gut colonization and yeast biofilm production capacity were investigated, by means of XTT reduction assay, in Clostridioides difficile infected (CDI) patients, in non-CDI diarrheic patients, and in healthy donors in two different time periods (2013-2015 and 2018-2019 respectively). Candida gut colonization was significantly (p < 0.001) associated to C. difficile infection, and to patients infected with hypervirulent C. difficile strains bearing the tcdC deletion at nucleotide 117 (p = 0.0003). Although there was not a prevalent yeast species in CDI patients, C. albicans was the species significantly (p < 0.001) associated to both the infections sustained by the non-hypervirulent C. difficile strains and those caused by the hypervirulent strain (p = 0.001). The biofilm production by the yeasts isolated from the CDI patients and from non-CDI diarrheic patients did not differ significantly. However, a significantly (p = 0.007) higher biofilm production was observed in the Candida strains, particularly C. albicans, isolated from healthy donors compared to that of the yeasts cultured from CDI patients. Seasonal occurrence was observed in the isolation rate of CDI and non-CDI diarrheic cases (p = 0.0019), peaking in winter for CDI patients and in spring for non-CDI diarrheic patients. Furthermore, seasonality emerged in the gut colonization by Candida of CDI patients in the winter. It seems, therefore, that the reduced capacity of biofilm production by Candida strains isolated from CDI patients might have a role in the development of C. difficile infection, probably facilitating the spread of the bacteria into the gut thus amplifying their pathogenic action.
Collapse
Affiliation(s)
- Grazia Brunetti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessandro Giuliani
- Department of Environment and Health, Istituto Superiore Di Sanità, Rome, Italy
| | - Anna Sara Navazio
- Department of Molecular Medicine, Laboratory of Microbiology and Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - Camilla Paradisi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Flavia Raponi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Libenzio Adrian Conti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
- Confocal Microscopy Core Facility, IRCSS Bambino Gesù Pediatric Hospital, Research Center, Rome, Italy
| | - Giammarco Raponi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
9
|
Zhang Q, Wang Y, Qu D, Yu J, Yang J. Role of HDAC6 inhibition in sepsis-induced acute respiratory distress syndrome (Review). Exp Ther Med 2021; 21:422. [PMID: 33747162 DOI: 10.3892/etm.2021.9866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) induced by sepsis contributes remarkably to the high mortality rate observed in intensive care units, largely due to a lack of effective drug therapies. Histone deacetylase 6 (HDAC6) is a class-IIb deacetylase that modulates non-nuclear protein functions via deacetylation and ubiquitination. Importantly, HDAC6 has been shown to exert anti-cancer, anti-neurodegeneration, and immunological effects, and several HDAC6 inhibitors have now entered clinical trials. It has also been recently shown to modulate inflammation, and HDAC6 inhibition has been demonstrated to markedly suppress experimental sepsis. The present review summarizes the role of HDAC6 in sepsis-induced inflammation and endothelial barrier dysfunction in recent years. It is proposed that HDAC6 inhibition predominantly ameliorates sepsis-induced ARDS by directly attenuating inflammation, which modulates the innate and adaptive immunity, transcription of pro-inflammatory genes, and protects endothelial barrier function. HDAC6 inhibition protects against sepsis-induced ARDS, thereby making HDAC6 a promising therapeutic target. However, HDAC inhibition may be associated with adverse effects on the embryo sac and oocyte, necessitating further studies.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Danhua Qu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Jinyan Yu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Junling Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
10
|
Youn GS, Park JK, Lee CY, Jang JH, Yun SH, Kwon HY, Choi SY, Park J. MicroRNA-22 negatively regulates LPS-induced inflammatory responses by targeting HDAC6 in macrophages. BMB Rep 2021. [PMID: 31964468 PMCID: PMC7196186 DOI: 10.5483/bmbrep.2020.53.4.209] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Dysregulation of histone deacetylase 6 (HDAC6) can lead to the pathologic states and result in the development of various diseases including cancers and inflammatory diseases. The objective of this study was to elucidate the regulatory role of microRNA-22 (miR-22) in HDAC6-mediated expression of proinflammatory cytokines in lipopolysaccharide (LPS)-stimulated macrophages. LPS stimulation induced HDAC6 expression, but suppressed miR-22 expression in macrophages, suggesting possible correlation between HDAC6 and miR-22. Luciferase reporter assays revealed that 3'UTR of HDAC6 was a bona fide target site of miR-22. Transfection of miR-22 mimic significantly inhibited LPS-induced HDAC6 expression, while miR-22 inhibitor further increased LPS-induced HDAC6 expression. LPS-induced activation of NF-κB and AP-1 was inhibited by miR-22 mimic, but further increased by miR-22 inhibitor. LPS-induced expression of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 was inhibited by miR-22 mimic, but further increased by miR-22 inhibitor. Taken together, these data provide evidence that miR-22 can downregulate LPS-induced expression of proinflammatory cytokines via suppression of NF-κB and AP-1 axis by targeting HDAC6 in macrophages. [BMB Reports 2020; 53(4): 223-228].
Collapse
Affiliation(s)
- Gi Soo Youn
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jong Kook Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Chae Yeon Lee
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jae Hee Jang
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Sang Ho Yun
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyeok Yil Kwon
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
11
|
Lin H, Fu G, Yu Q, Wang Z, Zuo Y, Shi Y, Zhang L, Gu Y, Qin L, Zhou T. Carbon black nanoparticles induce HDAC6-mediated inflammatory responses in 16HBE cells. Toxicol Ind Health 2020; 36:759-768. [PMID: 32783763 DOI: 10.1177/0748233720947214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Long-term inhalation of carbon black nanoparticles (CBNPs) leads to pulmonary inflammatory diseases. Histone deacetylase 6 (HDAC6) has been identified as an important regulator in the development of inflammatory disorders. However, the direct involvement of HDAC6 in CBNPs-induced pulmonary inflammatory responses remains unclear. To explore whether HDAC6 participates in CBNPs-induced pulmonary inflammation, human bronchial epithelial cell line (16HBE cells) was transfected with HDAC6 small interference RNA (siRNA) and then exposed to CBNPs at concentrations of 0, 25, and 50 µg/ml for 24 h. Intracellular HDAC6 and intraflagellar transport protein 88 (IFT88) mRNA and protein were determined by real-time polymerase chain reaction and Western blot, respectively. The secretions of inflammatory cytokines including interleukin (IL)-8, tumor necrosis factor (TNF)-α, IL-6, and IL-1β were measured by enzyme-linked immunosorbent assay. CBNPs induced a significant increase in the expressions of IL-8 and IL-6, accompanied by a high level of intracellular HDAC6 mRNA when compared with a blank control group (p < 0.05). However, there were no significant changes in the levels of TNF-α secretion, intracellular HDAC6 and IFT88 protein induced by CBNPs (p > 0.05). The HDAC6 mRNA expression was significantly suppressed in HDAC6 siRNA-transfected cells (p < 0.05). The secretions of IL-8, TNF-α, and IL-6 were significantly less in HDAC6 siRNA-transfected cells than that in normal 16HBE cells with exposure to 25 or 50 µg/ml of CBNPs, but intracellular IFT88 mRNA expression was markedly increased in HDAC6 siRNA-transfected cells when compared with normal 16HBE cells exposed to 50 µg/ml of CBNPs (all p < 0.05). Downregulation of the HDAC6 gene inhibits CBNPs-induced inflammatory responses in bronchial epithelial cells, partially through regulating IFT88 expression. It is suggested that CBNPs may trigger inflammatory responses in bronchial epithelial cells by an HDAC6/IFT88-dependent pathway.
Collapse
Affiliation(s)
- Hui Lin
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Guoqing Fu
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Qimei Yu
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zhenyu Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Basic Medicine, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yao Zuo
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yuqin Shi
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Ling Zhang
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yingying Gu
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Lingzhi Qin
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Medical College, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, 481115Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
12
|
Porcine Colostrum Protects the IPEC-J2 Cells and Piglet Colon Epithelium against Clostridioides (syn. Clostridium) difficile Toxin-Induced Effects. Microorganisms 2020; 8:microorganisms8010142. [PMID: 31968636 PMCID: PMC7022787 DOI: 10.3390/microorganisms8010142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/19/2019] [Accepted: 01/13/2020] [Indexed: 12/17/2022] Open
Abstract
Clostridioides difficile toxins are one of the main causative agents for the clinical symptoms observed during C. difficile infection in piglets. Porcine milk has been shown to strengthen the epithelial barrier function in the piglet’s intestine and may have the potential to neutralise clostridial toxins. We hypothesised that porcine colostrum exerts protective effects against those toxins in the IPEC-J2 cells and in the colon epithelium of healthy piglets. The IPEC-J2 cells were treated with either the toxins or porcine colostrum or their combination. Analyses included measurement of trans-epithelial electrical resistance (TEER), cell viability using propidium iodide by flow cytometry, gene expression of tight junction (TJ) proteins and immune markers, immunofluorescence (IF) histology of the cytoskeleton and a TJ protein assessment. Colon tissue explants from one- and two-week-old suckling piglets and from five-week-old weaned piglets were treated with C. difficile toxins in Ussing chamber assays to assess the permeability to macromolecules (FITC-dextran, HRP), followed by analysis of gene expression of TJ proteins and immune markers. Toxins decreased viability and integrity of IPEC-J2 cells in a time-dependent manner. Porcine colostrum exerted a protective effect against toxins as indicated by TEER and IF in IPEC-J2 cells. Toxins tended to increase paracellular permeability to macromolecules in colon tissues of two-week-old piglets and downregulated gene expression of occludin in colon tissues of five-week-old piglets (p = 0.05). Porcine milk including colostrum, besides other maternal factors, may be one of the important determinants of early immune programming towards protection from C. difficile infections in the offspring.
Collapse
|
13
|
Mucha J, Pawłowski TA, Klupczyńska EA, Guzicka M, Zadworny M. The Effect of Hydroxamic Siderophores Structure on Acetylation of Histone H3 and Alpha Tubulin in Pinus sylvestris Root Cells. Int J Mol Sci 2019; 20:E6099. [PMID: 31816938 PMCID: PMC6928989 DOI: 10.3390/ijms20236099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/18/2019] [Accepted: 11/30/2019] [Indexed: 12/03/2022] Open
Abstract
Protein acetylation affects gene expression, as well as other processes in cells, and it might be dependent on the availability of the metals. However, whether iron chelating compounds (siderophores) can have an effect on the acetylation process in plant roots is largely unknown. In the present study, western blotting and confocal microscopy was used to examine the degree of acetylation of histone H3 and alpha tubulin in Pinus sylvestris root cells in the presence of structurally different siderophores. The effect of metabolites that were produced by pathogenic and mycorrhizal fungi was also assessed. No effect was observed on histone acetylation. By contrast, the metabolites of the pathogenic fungus were able to decrease the level of microtubule acetylation, whereas treatment with iron-free ferrioxamine (DFO) was able to increase it. This latter was not observed when ferrioxamine-iron complexes were used. The pathogen metabolites induced important modifications of cytoskeleton organization. Siderophores also induced changes in the tubulin skeleton and these changes were iron-dependent. The effect of siderophores on the microtubule network was dependent on the presence of iron. More root cells with a depolymerized cytoskeleton were observed when the roots were exposed to iron-free siderophores and the metabolites of pathogenic fungi; whereas, the metabolites from mycorrhizal fungi and iron-enriched forms of siderophores slightly altered the cytoskeleton network of root cells. Collectively, these data indicated that the metabolites of pathogenic fungi mirror siderophore action, and iron limitation can lead to enhanced alternations in cell structure and physiology.
Collapse
Affiliation(s)
- Joanna Mucha
- Institute of Dendrology, Polish Academy of Sciences, Parkowa 5, 62-035 Kórnik, Poland; (T.A.P.); (E.A.K.); (M.G.); (M.Z.)
| | | | | | | | | |
Collapse
|
14
|
Zhang Y, Xie H, Tang W, Zeng X, Lin Y, Xu L, Xiao L, Xu J, Wu Z, Yuan D. Trichostatin A, a Histone Deacetylase Inhibitor, Alleviates Eosinophilic Meningitis Induced by Angiostrongylus cantonensis Infection in Mice. Front Microbiol 2019; 10:2280. [PMID: 31636619 PMCID: PMC6787401 DOI: 10.3389/fmicb.2019.02280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022] Open
Abstract
Histone deacetylase inhibitor (HDACi) has been used in the treatment of neurodegenerative or autoimmune diseases. Angiostrongyliasis cantonensis caused by Angiostrongylus cantonensis infection is an emerging zoonosis of human eosinophilic meningitis or meningoencephalitis. Progressive neuronal apoptosis is the pathological basis of behavioral dysfunctions in angiostrongyliasis cantonensis. Neurological defects after anthelmintic treatment for angiostrongyliasis cantonensis are still common. In this study, we examined the effects of trichostatin A (TSA), a HDACi, on eosinophilic meningitis induced by A. cantonensis in mice. Intragastric administration of TSA significantly ameliorated brain injury and decreased cognitive impairments in mice at 15 days post-infection. TSA administration effectively reduced the inflammatory factor levels of iNOS, TNF-α, IL-5, IL-6, and IL-13 in infected mice. TSA treatment counteracted apoptosis with reduced expression levels of cleaved caspase-3, -4, -6, and RIP3 in A. cantonensis infected mice. In addition, TSA administration reduced total HDAC activity and increased the acetylation of histone H3 and H4 in the brain tissue of infected mice. The underlying mechanism of TSA on eosinophilic meningitis might be associated with decreased NF-κB p65 nuclear accumulation by inhibiting IκB phosphorylation. Furthermore, a co-expressive network of NF-κB p65 with 22 other genes was constructed according to our previous transcriptomic data in infected mice. We identified the correlations in the gene expression of NF-κB p65 with Lrp10, Il12rb1, Nfkbia, Ube2n, and Ube2d1 in infected mice after TSA administration. Thus, TSA has a protective effect on the progression of eosinophilic meningitis induced by A. cantonensis in mice.
Collapse
Affiliation(s)
- Yanhua Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Hui Xie
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Wenyan Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xingda Zeng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Yu Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lian Xu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Lihua Xiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jun Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Dongjuan Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
15
|
Falcone M, Tiseo G, Venditti M, Menichetti F. Updates in the epidemiology and management of candidemia and Clostridioides difficile coinfection. Expert Rev Anti Infect Ther 2019; 17:375-382. [PMID: 30982376 DOI: 10.1080/14787210.2019.1608183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Introduction: In recent years, more and more studies have focused on the association between candidemia and Clostridioides difficile infection (CDI), highlighting the risk of subsequent candidemia in patients with CDI. However, a more recent model focuses on the Candida-Clostridioides difficile coinfection as a clinical entity in which candidemia could occur before or after the CDI episode. Areas covered: In this review we analyzed the physiopathological mechanisms underlying the Candida-Clostridioides difficile coinfection, both in case of candidemia preceding and following the CDI. We highlighted that gut alterations occurring during a CDI play a crucial role in the risk of subsequent candidemia. Moreover, we identified areas of interest about the management of Candida-Clostridioides difficile coinfection and proposed answers to relevant clinical questions. Expert opinion: The evaluation of risk factors for candidemia in patients with CDI and the rational antibiotic use in patients with candidemia remain the most efficacious and cost-free instruments to optimally manage the Candida-Clostridioides difficile coinfection. However, further studies are required to cover some unmet needs, such as the development of rapid diagnostic methods of candidemia and the use of new available drugs with minimal effect on the microbiome biodiversity in patients with CDI at high risk of fungemia.
Collapse
Affiliation(s)
- Marco Falcone
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Giusy Tiseo
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Mario Venditti
- b Department of Public Health and Infectious Diseases , 'Sapienza' University of Rome , Rome , Italy
| | - Francesco Menichetti
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
16
|
Inhibition of histone deacetylase 6 attenuates intestinal inflammation and apoptosis in a rodent model of hemorrhagic shock. J Trauma Acute Care Surg 2019; 86:874-880. [DOI: 10.1097/ta.0000000000002169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
17
|
Dynamic localization of α-tubulin acetyltransferase ATAT1 through the cell cycle in human fibroblastic KD cells. Med Mol Morphol 2018; 51:217-226. [PMID: 29869029 DOI: 10.1007/s00795-018-0195-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/01/2018] [Indexed: 12/22/2022]
Abstract
Acetylation of α-tubulin is a well-studied posttranscriptional modification, which is mostly catalyzed by α-tubulin N-acetyltransferase (ATAT1). ATAT1 possibly affects various cellular functions related with microtubules, such as intracellular transport, cell motility, cilia formation, and neuronal signaling. Here, we analyzed the subcellular localization of immunolabeled ATAT1 in human fibroblast KD cells through the cell cycle using confocal laser scanning microscopy. ATAT1 dramatically changed its localization through the cell cycle, depending on the mitotic phase. In interphase, immunolabeled ATAT1 was observed in centrioles, nuclei, and basal bodies if the cells projected primary cilia. ATAT1 was intensely detected as clusters in the nuclei in the G1-G2 phase. In telophase, ATAT1 colocalized with chromatids and spindle poles, and ultimately migrated to the daughter nucleus, newly synthesized centrioles, and midbody. The nucleolus is a core region of ribosomal RNA transcription, and the midbody is associated with severing and depolymerizing of microtubules in the stembody. The specific distributions of ATAT1 through the cell cycle suggest multiple functions of ATAT1, which could include acetylation of microtubules, RNA transcription activity, severing microtubules, and completion of cytokinesis.
Collapse
|
18
|
Wang J, Zhao L, Wei Z, Zhang X, Wang Y, Li F, Fu Y, Liu B. Inhibition of histone deacetylase reduces lipopolysaccharide-induced-inflammation in primary mammary epithelial cells by regulating ROS-NF-кB signaling pathways. Int Immunopharmacol 2018; 56:230-234. [PMID: 29414656 DOI: 10.1016/j.intimp.2018.01.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 01/15/2023]
Abstract
Histone deacetylase 6 (HDAC6) is the sole member of the HDAC family, that is predominantly located in the cytoplasm and has substrate specificity for nonhistone proteins, such as α-Tubulin. Although an increasing number of studies have shown that HDAC6 is involved in inflammatory diseases, but little is known about the participation of HDAC6 in the transcriptional regulation of inflammatory cytokines. Here, we examined the effects of Tubastatin (Tub), a highly selective HDAC6 inhibitor, on lipopolysaccharide (LPS)-stimulated primary bovine mammary epithelial cells (bMECs). The specific inhibition of HDAC6 using Tub significantly decreased the release of pro-inflammatory cytokines, such as TNF-α and IL-1β, which was associated with increased α-Tubulin acetylation. HDAC6 overexpression significantly induced reactive oxygen species (ROS) generation via upregulation of NADPH oxidase activity. Administration of Tub dose-dependently inhibited ROS production and NADPH oxidase activity. In addition, inhibition of HDAC6 led to suppression of the NF-κB signaling pathway. Thus, we report herein that HDAC6 is involved in ROS-NF-κB signaling pathway related to pro-inflammatory cytokine expression and that selective HDAC6 inhibition by Tub is a potent approach for preventing LPS-mediated inflammation.
Collapse
Affiliation(s)
- Jingjing Wang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Lilei Zhao
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Zhengkai Wei
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Xu Zhang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Yanan Wang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Yunhe Fu
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China; Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin Province 130062, People's Republic of China
| | - Bin Liu
- Cardiovascular disease center, First Hospital of Jilin University, Changchun, Jilin Province 130021, People's Republic of China.
| |
Collapse
|
19
|
Blockade of histone deacetylase 6 protects against cisplatin-induced acute kidney injury. Clin Sci (Lond) 2018; 132:339-359. [PMID: 29358506 DOI: 10.1042/cs20171417] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/04/2018] [Accepted: 01/22/2018] [Indexed: 12/21/2022]
Abstract
Histone deacetylase 6 (HDAC6) has been shown to be involved in various pathological conditions, including cancer, neurodegenerative disorders and inflammatory diseases. Nonetheless, its specific role in drug-induced nephrotoxicity is poorly understood. Cisplatin (dichlorodiamino platinum) belongs to an inorganic platinum - fundamental chemotherapeutic drug utilized in the therapy of various solid malignant tumors. However, the use of cisplatin is extremely limited by obvious side effects, for instance bone marrow suppression and nephrotoxicity. In the present study, we utilized a murine model of cisplatin-induced acute kidney injury (AKI) and a highly selective inhibitor of HDAC6, tubastatin A (TA), to assess the role of HDAC6 in nephrotoxicity and its associated mechanisms. Cisplatin-induced AKI was accompanied by increased expression and activation of HDAC6; blocking HDAC6 with TA lessened renal dysfunction, attenuated renal pathological changes, reduced expression of neutrophil gelatinase-associated lipocalin and kidney injury molecule 1, and decreased tubular cell apoptosis. In cultured human epithelial cells, TA or HDAC6 siRNA treatment also inhibited cisplatin-induced apoptosis. Mechanistic studies demonstrated that cisplatin treatment induced phosphorylation of AKT and loss of E-cadherin in the nephrotoxic kidney, and administration of TA enhanced AKT phosphorylation and preserved E-cadherin expression. HDAC6 inhibition also potentiated autophagy as evidenced by increased expression of autophagy-related gene (Atg) 7 (Atg7), Beclin-1, and decreased renal oxidative stress as demonstrated by up-regulation of superoxide dismutase (SOD) activity and down-regulation of malondialdehyde levels. Moreover, TA was effective in inhibiting nuclear factor-κ B (NF-κB) phosphorylation and suppressing the expression of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Collectively, these data provide strong evidence that HDAC6 inhibition is protective against cisplatin-induced AKI and suggest that HDAC6 may be a potential therapeutic target for AKI treatment.
Collapse
|
20
|
Zhang WB, Zhang HY, Jiao FZ, Wang LW, Zhang H, Gong ZJ. Histone deacetylase 6 inhibitor ACY-1215 protects against experimental acute liver failure by regulating the TLR4-MAPK/NF-κB pathway. Biomed Pharmacother 2017; 97:818-824. [PMID: 29112935 DOI: 10.1016/j.biopha.2017.10.103] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/20/2017] [Accepted: 10/21/2017] [Indexed: 02/06/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) is considered a new target for anticancer, anti-inflammatory, and neurodegenerative treatment. ACY-1215 is a selective histone deacetylase 6 inhibitor, and it has been recognized as a potential anticancer and anti-inflammation drug. The aim of our study was to investigate whether ACY-1215 has protective effects on acute liver failure (ALF) in mice and explore its potential mechanism. Male C57/BL6 mice were divided into normal, model, and ACY-1215 groups. ACY-1215 (25mg/kg) and same amounts of saline were given to mice. After 2h, the ALF models were induced by lipopolysaccharide (LPS, 100μg/kg) combined with D-galactosamine (D-gal, 400mg/kg). All animals were killed after 24h. The expressions of HDAC6 were determined by western blotting and RT-PCR assay. The expression levels of inflammatory cytokines were detected by ELISA and RT-PCR. The protein expression of Toll-like receptor 4 (TLR4), mitogen-activated protein kinase (MAPK), and nuclear factor κB (NF-κB) species were determined by western blot. The mortality of mice with ALF induced by LPS and D-gal was significantly decreased by ACY-1215 pretreatment. Procedures to manage ALF caused adversely affected liver histology and function; this damage was repaired by pretreatment of ACY-1215. ACY-1215 treatment also attenuated the serum and messenger RNA levels of the proinflammatory cytokines. Pretreatment of ACY-1215 significantly decreased the protein expression of TLR4 and the activation of MAPK and NF-κB signalling pathways. ACY-1215 has potential therapeutic value in mice with ALF by directly inhibiting inflammatory response via regulation of the TLR4-MAPK/NF-kB pathway.
Collapse
Affiliation(s)
- Wen-Bin Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Hai-Yue Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Fang-Zhou Jiao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Lu-Wen Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Hong Zhang
- Department of Pharmaceutical, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zuo-Jiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
21
|
Crosstalk between HDAC6 and Nox2-based NADPH oxidase mediates HIV-1 Tat-induced pro-inflammatory responses in astrocytes. Redox Biol 2017; 12:978-986. [PMID: 28499252 PMCID: PMC5429231 DOI: 10.1016/j.redox.2017.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 12/24/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) likely is important in inflammatory diseases. However, how HDAC6 exerts its effect on inflammatory processes remains unclear. HIV-1 transactivator of transcription (Tat) activates NADPH oxidase resulting in generation of reactive oxygen species (ROS), leading to extensive neuro-inflammation in the central nervous system. We investigated the correlation of HDAC6 and NADPH oxidase in HIV-1 Tat-stimulated astrocytes. HDAC6 knockdown attenuated HIV-1 Tat-induced ROS generation and NADPH oxidase activation. HDAC6 knockdown suppressed HIV-1 Tat-induced expression of NADPH oxidase subunits, such as Nox2, p47phox, and p22phox. Specific inhibition of HDAC6 using tubastatin A suppressed HIV-1 Tat-induced ROS generation and activation of NADPH oxidase. N-acetyl cysteine, diphenyl iodonium, and apocynin suppressed HIV-1 Tat-induced expression of HDAC6 and the pro-inflammatory chemokines CCL2, CXCL8, and CXCL10. Nox2 knockdown attenuated HIV-1 Tat-induced HDAC6 expression and subsequent expression of chemokines. The collective results point to the potential crosstalk between HDAC6 and NADPH oxidase, which could be a combined therapeutic target for relief of HIV-1 Tat-mediated neuro-inflammation. HDAC6 mediates HIV-1 Tat-induced ROS generation in astrocytes. HDAC6 is involved in HIV-1 Tat-induced activity and expression of Nox2-based NADPH oxidase. Crosstalk between HDAC6 and NADPH oxidase exists in HIV-1 Tat-stimulated astrocytes.
Collapse
|
22
|
Liu T, Wang R, Xu H, Song Y, Qi Y. A Highly Potent and Selective Histone Deacetylase 6 Inhibitor Prevents DSS-Induced Colitis in Mice. Biol Pharm Bull 2017; 40:936-940. [PMID: 28321036 DOI: 10.1248/bpb.b16-01023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammatory bowel disease (IBD) is a refractory illness with remarkably increasing incidence rate all over the world. However, no desirable treatment scheme is available. Therefore, research and development of new drugs for treating IBD are urgently needed. Histone deacetylase 6 (HDAC6) is considered to be a pro-inflammatory factor, thus the inhibitors specifically-targeting HDAC6 may find their way in IBD treatment. In this study, we evaluated the anti-inflammatory activity of a novel potent and selective HDAC6 inhibitor, LTB2, in dextran sulfate sodium (DSS)-induced colitis mouse model. It was found that LTB2 treatment significantly alleviated DSS-induced colitis in mice, as evidenced by body weight, colon length, histological examination, and the disease activity index (DAI) scores of rectal bleeding and diarrhea. More importantly, it showed a better protective effect on the DSS-induced colitis mice than the commonly used mesalazine in the clinic. Our results demonstrated that selective HDAC6 inhibitors may have a good prospect for IBD treatment.
Collapse
Affiliation(s)
- Ting Liu
- School of Pharmacy, Second Military Medical University
| | - Renping Wang
- School of Pharmacy, Second Military Medical University
| | - Haojie Xu
- School of Pharmacy, Second Military Medical University
| | - Yunlong Song
- School of Pharmacy, Second Military Medical University
| | - Yunpeng Qi
- School of Pharmacy, Second Military Medical University
| |
Collapse
|
23
|
Youn GS, Lee KW, Choi SY, Park J. Overexpression of HDAC6 induces pro-inflammatory responses by regulating ROS-MAPK-NF-κB/AP-1 signaling pathways in macrophages. Free Radic Biol Med 2016; 97:14-23. [PMID: 27208785 DOI: 10.1016/j.freeradbiomed.2016.05.014] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/29/2016] [Accepted: 05/16/2016] [Indexed: 11/24/2022]
Abstract
Although histone deacetylase 6 (HDAC6) has been implicated in inflammatory diseases, direct involvement and its action mechanism of HDAC6 in the transcriptional regulation of pro-inflammatory genes have been unclear. In this study, we investigated the possible role of HDAC6 in the expression of pro-inflammatory mediators, indicator of macrophage activation, in RAW 264.7 cells and primary mouse macrophages. HDAC6 overexpression significantly enhanced expression of pro-inflammatory cytokines, such as TNF-α, IL-1β, and IL-6, with concomitant reduction in acetylated α-tubulin. HDAC6 overexpression significantly induced ROS generation via upregulation of NADPH oxidase expression and activity. Inhibition of ROS generation by N-acetyl cysteine, diphenyl iodonium and apocynin suppressed HDAC6-induced pro-inflammatory cytokines. An HDAC6 enzymatic inhibitor significantly inhibited ROS generation and expression of HDAC6-induced pro-inflammatory mediators, indicating the requirement of HDAC6 enzymatic activity for induction of pro-inflammatory cytokines. In addition, HDAC6 overexpression increased activation of MAPK species including ERK, JNK, and p38. Furthermore, HDAC6 overexpression resulted in activation of the NF-κB and AP-1 signaling pathways. Overall, our results provide the first evidence that HDAC6 is capable of inducing expression of pro-inflammatory genes by regulating the ROS-MAPK-NF-κB/AP-1 pathways and serves as a molecular target for inflammation.
Collapse
Affiliation(s)
- Gi Soo Youn
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, 1 Hallymdaehak-gil, Chunchon 200-702, Kangwon-Do, Republic of Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, 1 Hallymdaehak-gil, Chunchon 200-702, Kangwon-Do, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, 1 Hallymdaehak-gil, Chunchon 200-702, Kangwon-Do, Republic of Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, 1 Hallymdaehak-gil, Chunchon 200-702, Kangwon-Do, Republic of Korea.
| |
Collapse
|
24
|
Lichtman JS, Ferreyra JA, Ng KM, Smits SA, Sonnenburg JL, Elias JE. Host-Microbiota Interactions in the Pathogenesis of Antibiotic-Associated Diseases. Cell Rep 2016; 14:1049-1061. [PMID: 26832403 PMCID: PMC5372703 DOI: 10.1016/j.celrep.2016.01.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 11/17/2015] [Accepted: 12/30/2015] [Indexed: 01/06/2023] Open
Abstract
Improved understanding of the interplay between host and microbes stands to illuminate new avenues for disease diagnosis, treatment, and prevention. Here, we provide a high-resolution view of the dynamics between host and gut microbiota during antibiotic-induced intestinal microbiota depletion, opportunistic Salmonella typhimurium and Clostridium difficile pathogenesis, and recovery from these perturbed states in a mouse model. Host-centric proteome and microbial community profiles provide a nuanced longitudinal view, revealing the interdependence between host and microbiota in evolving dysbioses. Time- and condition-specific molecular and microbial signatures are evident and clearly distinguished from pathogen-independent inflammatory fingerprints. Our data reveal that mice recovering from antibiotic treatment or C. difficile infection retain lingering signatures of inflammation, despite compositional normalization of the microbiota, and host responses could be rapidly and durably relieved through fecal transplant. These experiments demonstrate insights that emerge from the combination of these orthogonal, untargeted approaches to the gastrointestinal ecosystem.
Collapse
Affiliation(s)
- Joshua S Lichtman
- Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94025, USA
| | - Jessica A Ferreyra
- Department of Microbiology and Immunology, Stanford School of Medicine, Stanford University, Stanford, CA 94025, USA
| | - Katharine M Ng
- Department of Microbiology and Immunology, Stanford School of Medicine, Stanford University, Stanford, CA 94025, USA
| | - Samuel A Smits
- Department of Microbiology and Immunology, Stanford School of Medicine, Stanford University, Stanford, CA 94025, USA
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford School of Medicine, Stanford University, Stanford, CA 94025, USA
| | - Joshua E Elias
- Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford University, Stanford, CA 94025, USA.
| |
Collapse
|
25
|
Teng RJ, Wu TJ, Afolayan AJ, Konduri GG. Nitrotyrosine impairs mitochondrial function in fetal lamb pulmonary artery endothelial cells. Am J Physiol Cell Physiol 2016; 310:C80-C88. [PMID: 26491046 PMCID: PMC4698449 DOI: 10.1152/ajpcell.00073.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 10/19/2015] [Indexed: 12/16/2022]
Abstract
Nitration of both protein-bound and free tyrosine by reactive nitrogen species results in the formation of nitrotyrosine (NT). We previously reported that free NT impairs microtubule polymerization and uncouples endothelial nitric oxide synthase (eNOS) function in pulmonary artery endothelial cells (PAEC). Because microtubules modulate mitochondrial function, we hypothesized that increased NT levels during inflammation and oxidative stress will lead to mitochondrial dysfunction in PAEC. PAEC isolated from fetal lambs were exposed to varying concentrations of free NT. At low concentrations (1-10 μM), NT increased nitration of mitochondrial electron transport chain (ETC) protein subunit complexes I-V and state III oxygen consumption. Higher concentrations of NT (50 μM) caused decreased microtubule acetylation, impaired eNOS interactions with mitochondria, and decreased ETC protein levels. We also observed increases in heat shock protein-90 nitration, mitochondrial superoxide formation, and fragmentation of mitochondria in PAEC. Our data suggest that free NT accumulation may impair microtubule polymerization and exacerbate reactive oxygen species-induced cell damage by causing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Ru-Jeng Teng
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Tzong-Jin Wu
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Adeleye J Afolayan
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Girija G Konduri
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin
| |
Collapse
|
26
|
Li L, Yang XJ. Tubulin acetylation: responsible enzymes, biological functions and human diseases. Cell Mol Life Sci 2015; 72:4237-55. [PMID: 26227334 PMCID: PMC11113413 DOI: 10.1007/s00018-015-2000-5] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 07/22/2015] [Accepted: 07/24/2015] [Indexed: 12/28/2022]
Abstract
Microtubules have important functions ranging from maintenance of cell morphology to subcellular transport, cellular signaling, cell migration, and formation of cell polarity. At the organismal level, microtubules are crucial for various biological processes, such as viral entry, inflammation, immunity, learning and memory in mammals. Microtubules are subject to various covalent modifications. One such modification is tubulin acetylation, which is associated with stable microtubules and conserved from protists to humans. In the past three decades, this reversible modification has been studied extensively. In mammals, its level is mainly governed by opposing actions of α-tubulin acetyltransferase 1 (ATAT1) and histone deacetylase 6 (HDAC6). Knockout studies of the mouse enzymes have yielded new insights into biological functions of tubulin acetylation. Abnormal levels of this modification are linked to neurological disorders, cancer, heart diseases and other pathological conditions, thereby yielding important therapeutic implications. This review summarizes related studies and concludes that tubulin acetylation is important for regulating microtubule architecture and maintaining microtubule integrity. Together with detyrosination, glutamylation and other modifications, tubulin acetylation may form a unique 'language' to regulate microtubule structure and function.
Collapse
Affiliation(s)
- Lin Li
- Rosalind and Morris Goodman Cancer Research Center, Montreal, QC, H3A 1A3, Canada
- Department of Medicine, Montreal, QC, H3A 1A3, Canada
| | - Xiang-Jiao Yang
- Rosalind and Morris Goodman Cancer Research Center, Montreal, QC, H3A 1A3, Canada.
- Department of Medicine, Montreal, QC, H3A 1A3, Canada.
- Department of Biochemistry, McGill University, Montreal, QC, H3A 1A3, Canada.
- McGill University Health Center, Montreal, QC, H3A 1A3, Canada.
| |
Collapse
|
27
|
Nam ST, Hwang JH, Kim DH, Park MJ, Lee IH, Nam HJ, Kang JK, Kim SK, Hwang JS, Chung HK, Shong M, Lee CH, Kim H. Role of NADH: quinone oxidoreductase-1 in the tight junctions of colonic epithelial cells. BMB Rep 2015; 47:494-9. [PMID: 24393524 PMCID: PMC4206724 DOI: 10.5483/bmbrep.2014.47.9.196] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Indexed: 11/20/2022] Open
Abstract
NADH:quinone oxidoreductase 1 (NQO1) is known to be involved in the regulation of energy synthesis and metabolism, and the functional studies of NQO1 have largely focused on metabolic disorders. Here, we show for the first time that compared to NQO1-WT mice, NQO1-KO mice exhibited a marked increase of permeability and spontaneous inflammation in the gut. In the DSS-induced colitis model, NQO1-KO mice showed more severe inflammatory responses than NQO1-WT mice. Interestingly, the transcript levels of claudin and occludin, the major tight junction molecules of gut epithelial cells, were significantly decreased in NQO1-KO mice. The colons of NQO1-KO mice also showed high levels of reactive oxygen species (ROS) and histone deacetylase (HDAC) activity, which are known to affect transcriptional regulation. Taken together, these novel findings indicate that NQO1 contributes to the barrier function of gut epithelial cells by regulating the transcription of tight junction molecules.
Collapse
Affiliation(s)
- Seung Taek Nam
- Department of Life Science, College of Natural Science, Daejin University, Pocheon 487-711, Korea
| | - Jung Hwan Hwang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Korea
| | - Dae Hong Kim
- Department of Life Science, College of Natural Science, Daejin University, Pocheon 487-711, Korea
| | - Mi Jung Park
- Department of Life Science, College of Natural Science, Daejin University, Pocheon 487-711, Korea
| | - Ik Hwan Lee
- Department of Life Science, College of Natural Science, Daejin University, Pocheon 487-711, Korea
| | - Hyo Jung Nam
- Department of Life Science, College of Natural Science, Daejin University, Pocheon 487-711, Korea
| | - Jin Ku Kang
- Department of Life Science, College of Natural Science, Daejin University, Pocheon 487-711, Korea
| | - Sung Kuk Kim
- Department of Life Science, College of Natural Science, Daejin University, Pocheon 487-711, Korea
| | - Jae Sam Hwang
- Department of Agricultural Biology, National Academy of Agricultural Science, RDA, Suwon 441-707, Korea
| | - Hyo Kyun Chung
- Department of Internal Medicine, Chungnam National University, Daejon 301-721, Korea
| | - Minho Shong
- Department of Internal Medicine, Chungnam National University, Daejon 301-721, Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Korea
| | - Ho Kim
- Department of Internal Medicine, Chungnam National University, Daejon 301-721, Korea
| |
Collapse
|
28
|
|
29
|
Russo A, Falcone M, Fantoni M, Murri R, Masucci L, Carfagna P, Ghezzi MC, Posteraro B, Sanguinetti M, Venditti M. Risk factors and clinical outcomes of candidaemia in patients treated for Clostridium difficile infection. Clin Microbiol Infect 2015; 21:493.e1-4. [PMID: 25698658 DOI: 10.1016/j.cmi.2014.12.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/12/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Abstract
The alterations occurring in the intestinal flora during Clostridium difficile infection (CDI) may promote the translocation of Candida to the blood and the development of candidaemia. The aim of our study was to analyse clinical findings of these patients to determine the risk factors associated with the development of candidaemia subsequent to CDI. We compared 35 patients with candidaemia subsequent to CDI with 105 patients with CDI. Patients with candidaemia showed more severe infections and higher mortality. The ribotype 027 strain and vancomycin treatment at ≥ 1000 mg/day were prevalent in patients developing candidaemia. CDI may predispose to the translocation of Candida.
Collapse
Affiliation(s)
- A Russo
- Department of Public Health and Infectious Diseases, 'Sapienza' University of Rome, Italy
| | - M Falcone
- Department of Public Health and Infectious Diseases, 'Sapienza' University of Rome, Italy
| | - M Fantoni
- Institute of Infectious Diseases, Università Cattolica del Sacro Cuore, Italy
| | - R Murri
- Institute of Infectious Diseases, Università Cattolica del Sacro Cuore, Italy
| | - L Masucci
- Institute of Microbiology, Università Cattolica del Sacro Cuore, Italy
| | - P Carfagna
- 'San Giovanni Addolorata' Hospital, Italy
| | - M C Ghezzi
- Department of Public Health and Infectious Diseases, 'Sapienza' University of Rome, Italy
| | - B Posteraro
- Institute of Public Health (Section of Hygiene), Università Cattolica del Sacro Cuore, Rome, Italy
| | - M Sanguinetti
- Institute of Microbiology, Università Cattolica del Sacro Cuore, Italy
| | - M Venditti
- Department of Public Health and Infectious Diseases, 'Sapienza' University of Rome, Italy.
| |
Collapse
|
30
|
Critical roles of Clostridium difficile toxin B enzymatic activities in pathogenesis. Infect Immun 2014; 83:502-13. [PMID: 25404023 DOI: 10.1128/iai.02316-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
TcdB is one of the key virulence factors of Clostridium difficile that is responsible for causing serious and potentially fatal colitis. The toxin contains at least two enzymatic domains: an effector glucosyltransferase domain for inactivating host Rho GTPases and a cysteine protease domain for the delivery of the effector domain into host cytosol. Here, we describe a novel intrabody approach to examine the role of these enzymes of TcdB in cellular intoxication. By screening a single-domain heavy chain (V(H)H) library raised against TcdB, we identified two V(H)H antibodies, 7F and E3, that specifically inhibit TcdB cysteine protease and glucosyltransferase activities, respectively. Cytoplasmic expression of 7F intrabody in Vero cells inhibited TcdB autoprocessing and delayed cellular intoxication, whereas E3 intrabody completely blocked the cytopathic effects of TcdB holotoxin. These data also demonstrate for the first time that toxin autoprocessing occurs after cysteine protease and glucosyltransferase domains translocate into the cytosol of target cells. We further determined the role of the enzymatic activities of TcdB in in vivo toxicity using a sensitive systemic challenge model in mice. Consistent with these in vitro results, a cysteine protease noncleavable mutant, TcdB-L543A, delayed toxicity in mice, whereas glycosyltransferase-deficient TcdB demonstrated no toxicity up to 500-fold of the 50% lethal dose (LD50) when it was injected systemically. Thus, glucosyltransferase but not cysteine protease activity is critical for TcdB-mediated cytopathic effects and TcdB systemic toxicity, highlighting the importance of targeting toxin glucosyltransferase activity for future therapy.
Collapse
|
31
|
Persistence and toxin production by Clostridium difficile within human intestinal organoids result in disruption of epithelial paracellular barrier function. Infect Immun 2014; 83:138-45. [PMID: 25312952 DOI: 10.1128/iai.02561-14] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Clostridium difficile is the leading cause of infectious nosocomial diarrhea. The pathogenesis of C. difficile infection (CDI) results from the interactions between the pathogen, intestinal epithelium, host immune system, and gastrointestinal microbiota. Previous studies of the host-pathogen interaction in CDI have utilized either simple cell monolayers or in vivo models. While much has been learned by utilizing these approaches, little is known about the direct interaction of the bacterium with a complex host epithelium. Here, we asked if human intestinal organoids (HIOs), which are derived from pluripotent stem cells and demonstrate small intestinal morphology and physiology, could be used to study the pathogenesis of the obligate anaerobe C. difficile. Vegetative C. difficile, microinjected into the lumen of HIOs, persisted in a viable state for up to 12 h. Upon colonization with C. difficile VPI 10463, the HIO epithelium is markedly disrupted, resulting in the loss of paracellular barrier function. Since similar effects were not observed when HIOs were colonized with the nontoxigenic C. difficile strain F200, we directly tested the role of toxin using TcdA and TcdB purified from VPI 10463. We show that the injection of TcdA replicates the disruption of the epithelial barrier function and structure observed in HIOs colonized with viable C. difficile.
Collapse
|
32
|
Li Y, Hu X, Zhang X, Liu Z, Ding X, Xia L, Hu S. Photorhabdus luminescensPirAB-fusion protein exhibits both cytotoxicity and insecticidal activity. FEMS Microbiol Lett 2014; 356:23-31. [DOI: 10.1111/1574-6968.12474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/29/2014] [Accepted: 05/07/2014] [Indexed: 10/25/2022] Open
Affiliation(s)
- Yusheng Li
- College of Life Sciences; Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology; Hunan Normal University; Changsha China
| | - Xiaofeng Hu
- College of Life Sciences; Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology; Hunan Normal University; Changsha China
| | - Xu Zhang
- College of Life Sciences; Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology; Hunan Normal University; Changsha China
| | - Zhengqiang Liu
- College of Life Sciences; Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology; Hunan Normal University; Changsha China
| | - Xuezhi Ding
- College of Life Sciences; Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology; Hunan Normal University; Changsha China
| | - Liqiu Xia
- College of Life Sciences; Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology; Hunan Normal University; Changsha China
| | - Shengbiao Hu
- College of Life Sciences; Hunan Provincial Key Laboratory of Microbial Molecular Biology - State Key Laboratory Breeding Base of Microbial Molecular Biology; Hunan Normal University; Changsha China
| |
Collapse
|
33
|
Busbee PB, Nagarkatti M, Nagarkatti PS. Natural indoles, indole-3-carbinol and 3,3'-diindolymethane, inhibit T cell activation by staphylococcal enterotoxin B through epigenetic regulation involving HDAC expression. Toxicol Appl Pharmacol 2014; 274:7-16. [PMID: 24200994 PMCID: PMC3874587 DOI: 10.1016/j.taap.2013.10.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 12/13/2022]
Abstract
Staphylococcal enterotoxin B (SEB) is a potent exotoxin produced by the Staphylococcus aureus. This toxin is classified as a superantigen because of its ability to directly bind with MHC-II class molecules followed by activation of a large proportion of T cells bearing specific Vβ-T cell receptors. Commonly associated with classic food poisoning, SEB has also been shown to induce toxic shock syndrome, and is also considered to be a potential biological warfare agent because it is easily aerosolized. In the present study, we assessed the ability of indole-3-carbinol (I3C) and one of its byproducts, 3,3'-diindolylmethane (DIM), found in cruciferous vegetables, to counteract the effects of SEB-induced activation of T cells in mice. Both I3C and DIM were found to decrease the activation, proliferation, and cytokine production by SEB-activated Vβ8(+) T cells in vitro and in vivo. Interestingly, inhibitors of histone deacetylase class I (HDAC-I), but not class II (HDAC-II), showed significant decrease in SEB-induced T cell activation and cytokine production, thereby suggesting that epigenetic modulation plays a critical role in the regulation of SEB-induced inflammation. In addition, I3C and DIM caused a decrease in HDAC-I but not HDAC-II in SEB-activated T cells, thereby suggesting that I3C and DIM may inhibit SEB-mediated T cell activation by acting as HDAC-I inhibitors. These studies not only suggest for the first time that plant-derived indoles are potent suppressors of SEB-induced T cell activation and cytokine storm but also that they may mediate these effects by acting as HDAC inhibitors.
Collapse
Affiliation(s)
- Philip B Busbee
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| |
Collapse
|
34
|
Zhang X, Hu X, Li Y, Ding X, Yang Q, Sun Y, Yu Z, Xia L, Hu S. XaxAB-like binary toxin from Photorhabdus luminescens exhibits both insecticidal activity and cytotoxicity. FEMS Microbiol Lett 2013; 350:48-56. [PMID: 24188660 DOI: 10.1111/1574-6968.12321] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/11/2013] [Accepted: 10/30/2013] [Indexed: 11/26/2022] Open
Abstract
The enterobacterium Photorhabdus luminescens produces a number of toxins to kill its insect host. By analyzing the genomic sequence of P. luminescens TT01, we found that amino acid sequences encoded by plu1961 and plu1962 showed high similarity to XaxAB binary toxin of Xenorhabuds nematophila, which has both necrotic and apoptotic activities in both insect and mammalian cells in vitro. To evaluate the biological activity of Plu1961/Plu1962, their coding genes were cloned and expressed in Escherichia coli. Both Plu1961 and Plu1962 were expressed as soluble protein in BL21 (DE3) and their mixture caused insect midgut CF-203 cells death via necrosis. Confocal fluorescence microscopy showed that Plu1961/Plu1962 mixture was able to depolymerize microtubule and induce the increase in plasma membrane permeabilization in CF-203 cells. Moreover, co-expression of Plu1961/Plu1962 in the same cytoplasm exhibited cytotoxic effect against mammalian cells (B16, 4T1, and HeLa cells) and injectable activity against Spodoptera exigua larvae. Until now, two types of binary toxins have been identified in P. luminescens, the first type is PirAB and Plu1961/Plu1962 is the second one. The biological role of Plu1961/Plu1962 binary toxin played in the infection process should attract more attention in future.
Collapse
Affiliation(s)
- Xu Zhang
- College of Life Science, Hunan Normal University, Hunan Provincial Key Laboratory of Microbial Molecular Biology-State Key Laboratory Breeding Base of Microbial Molecular Biology, Changsha, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhang D, Wu CT, Qi X, Meijering RAM, Hoogstra-Berends F, Tadevosyan A, Cubukcuoglu Deniz G, Durdu S, Akar AR, Sibon OCM, Nattel S, Henning RH, Brundel BJJM. Activation of histone deacetylase-6 induces contractile dysfunction through derailment of α-tubulin proteostasis in experimental and human atrial fibrillation. Circulation 2013; 129:346-58. [PMID: 24146251 DOI: 10.1161/circulationaha.113.005300] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is characterized by structural remodeling, contractile dysfunction, and AF progression. Histone deacetylases (HDACs) influence acetylation of both histones and cytosolic proteins, thereby mediating epigenetic regulation and influencing cell proteostasis. Because the exact function of HDACs in AF is unknown, we investigated their role in experimental and clinical AF models. METHODS AND RESULTS Tachypacing of HL-1 atrial cardiomyocytes and Drosophila pupae hearts significantly impaired contractile function (amplitude of Ca(2+) transients and heart wall contractions). This dysfunction was prevented by inhibition of HDAC6 (tubacin) and sirtuins (nicotinamide). Tachypacing induced specific activation of HDAC6, resulting in α-tubulin deacetylation, depolymerization, and degradation by calpain. Tachypacing-induced contractile dysfunction was completely rescued by dominant-negative HDAC6 mutants with loss of deacetylase activity in the second catalytic domain, which bears α-tubulin deacetylase activity. Furthermore, in vivo treatment with the HDAC6 inhibitor tubastatin A protected atrial tachypaced dogs from electric remodeling (action potential duration shortening, L-type Ca(2+) current reduction, AF promotion) and cellular Ca(2+)-handling/contractile dysfunction (loss of Ca(2+) transient amplitude, sarcomere contractility). Finally, atrial tissue from patients with AF also showed a significant increase in HDAC6 activity and reduction in the expression of both acetylated and total α-tubulin. CONCLUSIONS AF induces remodeling and loss of contractile function, at least in part through HDAC6 activation and subsequent derailment of α-tubulin proteostasis and disruption of the cardiomyocyte microtubule structure. In vivo inhibition of HDAC6 protects against AF-related atrial remodeling, disclosing the potential of HDAC6 as a therapeutic target in clinical AF.
Collapse
Affiliation(s)
- Deli Zhang
- Departments of Clinical Pharmacology (D.Z., R.A.M.M., F.H.-B., R.H.H., B.J.J.M.B.) and Cell Biology (O.C.M.S.), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Research Center and Department of Medicine, Montreal Heart Institute and Université de Montréal, Montreal, QB, Canada (C.T.W., X.Y.Q., A.T., S.N.); Chang-Gung Memorial Hospital and Chang-Gung University, Taoyuan, Taiwan, Republic of China (C.T.W.); Nyken BV, Groningen, The Netherlands (F.-H.B.); Ankara University Biotechnology Institute, Ankara, Turkey (G.C.D., S.D.); Ankara University Stem Cell Institute, Ankara, Turkey (G.C.D., S.D., A.R.A.); Department of Cardiovascular Surgery, Ankara University School of Medicine, Ankara, Turkey (S.D., A.R.A.); and Department of Pharmacology, McGill University, Montreal, QB, Canada (S.N.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hing TC, Ho S, Shih DQ, Ichikawa R, Cheng M, Chen J, Chen X, Law I, Najarian R, Kelly CP, Gallo RL, Targan SR, Pothoulakis C, Koon HW. The antimicrobial peptide cathelicidin modulates Clostridium difficile-associated colitis and toxin A-mediated enteritis in mice. Gut 2013; 62:1295-305. [PMID: 22760006 PMCID: PMC3737259 DOI: 10.1136/gutjnl-2012-302180] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Clostridium difficile mediates intestinal inflammation by releasing toxin A (TxA), a potent enterotoxin. Cathelicidins (Camp as gene name, LL-37 peptide in humans and mCRAMP peptide in mice) are antibacterial peptides that also posses anti-inflammatory properties. OBJECTIVES To determine the role of cathelicidins in models of Clostridium difficile infection and TxA-mediated ileal inflammation and cultured human primary monocytes. DESIGN Wild-type (WT) and mCRAMP-deficient (Camp(-/-)) mice were treated with an antibiotic mixture and infected orally with C difficile. Some mice were intracolonically given mCRAMP daily for 3 days. Ileal loops were also prepared in WT mice and treated with either saline or TxA and incubated for 4 h, while some TxA-treated loops were injected with mCRAMP. RESULTS Intracolonic mCRAMP administration to C difficile-infected WT mice showed significantly reduced colonic histology damage, apoptosis, tissue myeloperoxidase (MPO) and tumour necrosis factor (TNF)α levels. Ileal mCRAMP treatment also significantly reduced histology damage, tissue apoptosis, MPO and TNFα levels in TxA-exposed ileal loops. WT and Camp(-/-) mice exhibited similar intestinal responses in both models, implying that C difficile/TxA-induced endogenous cathelicidin may be insufficient to modulate C difficile/TxA-mediated intestinal inflammation. Both LL-37 and mCRAMP also significantly reduced TxA-induced TNFα secretion via inhibition of NF-κB phosphorylation. Endogenous cathelicidin failed to control C difficile and/or toxin A-mediated inflammation and even intestinal cathelicidin expression was increased in humans and mice. CONCLUSION Exogenous cathelicidin modulates C difficile colitis by inhibiting TxA-associated intestinal inflammation. Cathelicidin administration may be a new anti-inflammatory treatment for C difficile toxin-associated disease.
Collapse
Affiliation(s)
- Tressia C Hing
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| | - Samantha Ho
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| | - David Q Shih
- Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Ryan Ichikawa
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| | - Michelle Cheng
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| | - Jeremy Chen
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy Law
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| | - Robert Najarian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ciaran P Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard L Gallo
- Division of Dermatology, the University of California San Diego, San Diego, California, USA
| | - Stephan R Targan
- Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Charalabos Pothoulakis
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| | - Hon Wai Koon
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, the University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
37
|
In vivo physiological and transcriptional profiling reveals host responses to Clostridium difficile toxin A and toxin B. Infect Immun 2013; 81:3814-24. [PMID: 23897615 DOI: 10.1128/iai.00869-13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Toxin A (TcdA) and toxin B (TcdB) of Clostridium difficile cause gross pathological changes (e.g., inflammation, secretion, and diarrhea) in the infected host, yet the molecular and cellular pathways leading to observed host responses are poorly understood. To address this gap, we evaluated the effects of single doses of TcdA and/or TcdB injected into the ceca of mice, and several endpoints were analyzed, including tissue pathology, neutrophil infiltration, epithelial-layer gene expression, chemokine levels, and blood cell counts, 2, 6, and 16 h after injection. In addition to confirming TcdA's gross pathological effects, we found that both TcdA and TcdB resulted in neutrophil infiltration. Bioinformatics analyses identified altered expression of genes associated with the metabolism of lipids, fatty acids, and detoxification; small GTPase activity; and immune function and inflammation. Further analysis revealed transient expression of several chemokines (e.g., Cxcl1 and Cxcl2). Antibody neutralization of CXCL1 and CXCL2 did not affect TcdA-induced local pathology or neutrophil infiltration, but it did decrease the peripheral blood neutrophil count. Additionally, low serum levels of CXCL1 and CXCL2 corresponded with greater survival. Although TcdA induced more pronounced transcriptional changes than TcdB and the upregulated chemokine expression was unique to TcdA, the overall transcriptional responses to TcdA and TcdB were strongly correlated, supporting differences primarily in timing and potency rather than differences in the type of intracellular host response. In addition, the transcriptional data revealed novel toxin effects (e.g., altered expression of GTPase-associated and metabolic genes) underlying observed physiological responses to C. difficile toxins.
Collapse
|
38
|
Guastalegname M, Russo A, Falcone M, Giuliano S, Venditti M. Candidemia subsequent to severe infection due to Clostridium difficile: is there a link? Clin Infect Dis 2013; 57:772-4. [PMID: 23723197 DOI: 10.1093/cid/cit362] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
39
|
Kim GW, Li L, Ghorbani M, You L, Yang XJ. Mice lacking α-tubulin acetyltransferase 1 are viable but display α-tubulin acetylation deficiency and dentate gyrus distortion. J Biol Chem 2013; 288:20334-50. [PMID: 23720746 DOI: 10.1074/jbc.m113.464792] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
α-Tubulin acetylation at Lys-40, located on the luminal side of microtubules, has been widely studied and used as a marker for stable microtubules in the cilia and other subcellular structures, but the functional consequences remain perplexing. Recent studies have shown that Mec-17 and its paralog are responsible for α-tubulin acetylation in Caenorhabditis elegans. There is one such protein known as Atat1 (α-tubulin acetyltransferase 1) per higher organism. Zebrafish Atat1 appears to govern embryo development, raising the intriguing possibility that Atat1 is also critical for development in mammals. In addition to Atat1, three other mammalian acetyltransferases, ARD1-NAT1, ELP3, and GCN5, have been shown to acetylate α-tubulin in vitro, so an important question is how these four enzymes contribute to the acetylation in vivo. We demonstrate here that Atat1 is a major α-tubulin acetyltransferase in mice. It is widely expressed in mouse embryos and tissues. Although Atat1-null animals display no overt phenotypes, α-tubulin acetylation is lost in sperm flagella and the dentate gyrus is slightly deformed. Furthermore, human ATAT1 colocalizes on bundled microtubules with doublecortin. These results thus suggest that mouse Atat1 may regulate advanced functions such as learning and memory, thereby shedding novel light on the physiological roles of α-tubulin acetylation in mammals.
Collapse
Affiliation(s)
- Go-Woon Kim
- Rosalind and Morris Goodman Cancer Research Center, Montréal, Québec H3A 1A3, Canada
| | | | | | | | | |
Collapse
|
40
|
Glutamine and alanyl-glutamine increase RhoA expression and reduce Clostridium difficile toxin-a-induced intestinal epithelial cell damage. BIOMED RESEARCH INTERNATIONAL 2012; 2013:152052. [PMID: 23484083 PMCID: PMC3591182 DOI: 10.1155/2013/152052] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/12/2012] [Indexed: 12/18/2022]
Abstract
Clostridium difficile is a major cause of antibiotic-associated colitis and is associated with significant morbidity and mortality. Glutamine (Gln) is a major fuel for the intestinal cell population. Alanyl-glutamine (Ala-Gln) is a dipeptide that is highly soluble and well tolerated. IEC-6 cells were used in the in vitro experiments. Cell morphology was evaluated by atomic force microscopy (AFM) and scanning electron microscopy (SEM). Cell proliferation was assessed by WST-1 and Ki-67 and apoptosis was assessed by TUNEL. Cytoskeleton was evaluated by immunofluorescence for RhoA and F-actin. RhoA was quantified by immunoblotting. TcdA induced cell shrinkage as observed by AFM, SEM, and fluorescent microscopy. Additionally, collapse of the F-actin cytoskeleton was demonstrated by immunofluorescence. TcdA decreased cell volume and area and increased cell height by 79%, 66.2%, and 58.9%, respectively. Following TcdA treatment, Ala-Gln and Gln supplementation, significantly increased RhoA by 65.5% and 89.7%, respectively at 24 h. Ala-Gln supplementation increased cell proliferation by 137.5% at 24 h and decreased cell apoptosis by 61.4% at 24 h following TcdA treatment. In conclusion, TcdA altered intestinal cell morphology and cytoskeleton organization, decreased cell proliferation, and increased cell apoptosis. Ala-Gln and Gln supplementation reduced intestinal epithelial cell damage and increased RhoA expression.
Collapse
|
41
|
Roberts A, McGlashan J, Al-Abdulla I, Ling R, Denton H, Green S, Coxon R, Landon J, Shone C. Development and evaluation of an ovine antibody-based platform for treatment of Clostridium difficile infection. Infect Immun 2012; 80:875-82. [PMID: 22144483 PMCID: PMC3264293 DOI: 10.1128/iai.05684-11] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 11/18/2011] [Indexed: 12/18/2022] Open
Abstract
Treatment of Clostridium difficile is a major problem as a hospital-associated infection which can cause severe, recurrent diarrhea. The currently available antibiotics are not effective in all cases and alternative treatments are required. In the present study, an ovine antibody-based platform for passive immunotherapy of C. difficile infection is described. Antibodies with high toxin-neutralizing titers were generated against C. difficile toxins A and B and were shown to neutralize three sequence variants of these toxins (toxinotypes) which are prevalent in human C. difficile infection. Passive immunization of hamsters with a mixture of toxin A and B antibodies protected them from a challenge with C. difficile spores in a dose-dependent manner. Antibodies to both toxins A and B were required for protection. The administration of toxin A and B antibodies up to 24 h postchallenge was found to reduce significantly the onset of C. difficile infection compared to nonimmunized controls. Protection from infection was also demonstrated with key disease isolates (ribotypes 027 and 078), which are members of the hypervirulent C. difficile clade. The ribotype 027 and 078 strains also have the capacity to produce an active binary toxin and these data suggest that neutralization of this toxin is unnecessary for the management of infection induced by these strains. In summary, the data suggest that ovine toxin A and B antibodies may be effective in the treatment of C. difficile infection; their potential use for the management of severe, fulminant cases is discussed.
Collapse
Affiliation(s)
- April Roberts
- Health Protection Agency, Porton Down, Salisbury, Wilts, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Shen A. Clostridium difficile toxins: mediators of inflammation. J Innate Immun 2012; 4:149-58. [PMID: 22237401 DOI: 10.1159/000332946] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 09/06/2011] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile is a significant problem in hospital settings as the most common cause of nosocomial diarrhea worldwide. C. difficile infections (CDIs) are characterized by an acute intestinal inflammatory response with neutrophil infiltration. These symptoms are primarily caused by the glucosylating toxins, TcdA and TcdB. In the past decade, the frequency and severity of CDIs have increased markedly due to the emergence of so-called hypervirulent strains that overproduce cytotoxic glucosylating toxins relative to historical strains. In addition, these strains produce a third toxin, binary toxin or C. difficile transferase (CDT), that may contribute to hypervirulence. Both the glucosylating toxins and CDT covalently modify target cell proteins to cause disassembly of the actin cytoskeleton and induce severe inflammation. This review summarizes our current knowledge of the mechanisms by which glucosylating toxins and CDT disrupt target cell function, alter host physiology and stimulate immune responses.
Collapse
Affiliation(s)
- Aimee Shen
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, Vt. 05401, USA.
| |
Collapse
|
43
|
Popoff MR. Multifaceted interactions of bacterial toxins with the gastrointestinal mucosa. Future Microbiol 2011; 6:763-97. [PMID: 21797691 DOI: 10.2217/fmb.11.58] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The digestive tract is one of the ecosystems that harbors the largest number and greatest variety of bacteria. Among them, certain bacteria have developed various strategies, including the synthesis of virulence factors such as toxins, to interact with the intestinal mucosa, and are responsible for various pathologies. A large variety of bacterial toxins of different sizes, structures and modes of action are able to interact with the gastrointestinal mucosa. Some toxins, termed enterotoxins, directly stimulate fluid secretion in enterocytes or cause their death, whereas other toxins pass through the intestinal barrier and disseminate by the general circulation to remote organs or tissues, where they are active. After recognition of a membrane receptor on target cells, toxins can act at the cell membrane by transducing a signal across the membrane in a hormone-like manner, by pore formation or by damaging membrane compounds. Other toxins can enter the cells and modify an intracellular target leading to a disregulation of certain physiological processes or disorganization of some structural architectures and cell death. Toxins are fascinating molecules, which mimic or interfere with eukaryotic physiological processes. Thereby, they have permitted the identification and characterization of new natural hormones or regulatory pathways. Besides use as protective antigens in vaccines, toxins offer multiple possibilities in pharmacology, such as immune modulation or specific delivery of a protein of interest into target cells.
Collapse
Affiliation(s)
- M R Popoff
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, 25 rue du Dr Roux, 757245 Paris cedex 15, France.
| |
Collapse
|
44
|
Identification of a novel virulence factor in Clostridium difficile that modulates toxin sensitivity of cultured epithelial cells. Infect Immun 2011; 79:3810-20. [PMID: 21746858 DOI: 10.1128/iai.00051-11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Two glucosylating toxins named toxins A and B play a role in the pathogenesis of Clostridium Difficile infection. The interaction of the toxins with host cell factors proceeds to downstream stages of cytotoxic effects in cells, in which involvement of other C. difficile factors remains unknown. We utilized culture filtrate of C. difficile with a low dilution to characterize the influence of putative minor proteins on the organization of the actin cytoskeleton in cultured epithelial cells and found a previously uncharacterized F-actin aggregated structure, termed "actin aggregate," at the juxtanuclear region. We reasoned that formation of actin aggregate was due to an additional factor(s) in the culture filtrate rather than the glucosylating toxins, because treatment of purified toxins rarely caused actin aggregate in cells. We focused on a previously uncharacterized hypothetical protein harboring a KDEL-like sequence as a candidate. The product of the candidate gene was detected in culture filtrate of C. difficile ATCC 9689 and was renamed Srl. Purified glutathione S-transferase-tagged Srl triggered formation of actin aggregate in the cells in the presence of either toxin A or B and enhanced cytotoxicity of each of the two toxins, including decreases in both cell viability and transepithelial resistance of cultured epithelial monolayer, although the recombinant Srl alone did not show detectable cytotoxicity. Srl-neutralized culture filtrate partially inhibited morphological changes of the cells in parallel with decreased actin aggregate formation in the cells. Thus, Srl might contribute to the modulation of toxin sensitivity of intestinal epithelial cells by enhancing cytotoxicity of C. difficile toxins.
Collapse
|
45
|
Popoff MR, Geny B. Rho/Ras-GTPase-dependent and -independent activity of clostridial glucosylating toxins. J Med Microbiol 2011; 60:1057-1069. [PMID: 21349986 DOI: 10.1099/jmm.0.029314-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Clostridial glucosylating toxins are the main virulence factors of clostridia responsible for gangrene and/or colitis. These toxins have been well characterized to inactivate Rho/Ras-GTPases through glucosylation. However, the signalling pathways downstream of Rho/Ras-GTPases leading to the intracellular effects of these toxins are only partially known. Rac-dependent modification of focal adhesion complexes and phosphoinositide metabolism seem to be key processes involved in actin filament depolymerization and disorganization of intercellular junctions. In addition, clostridial glucosylating toxins induce Rho/Ras-independent intracellular effects such as activation of mitogen-activated protein kinase pathways, which are used by some of these toxins to trigger an inflammatory response.
Collapse
Affiliation(s)
- Michel R Popoff
- Institut Pasteur, Unité des Bactéries Anaérobies et Toxines, 25 rue du Dr Roux, 75724 Paris cedex 15, France
| | - Bladine Geny
- Institut Pasteur, Unité des Bactéries Anaérobies et Toxines, 25 rue du Dr Roux, 75724 Paris cedex 15, France
| |
Collapse
|
46
|
Perdiz D, Mackeh R, Poüs C, Baillet A. The ins and outs of tubulin acetylation: more than just a post-translational modification? Cell Signal 2010; 23:763-71. [PMID: 20940043 DOI: 10.1016/j.cellsig.2010.10.014] [Citation(s) in RCA: 171] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 10/01/2010] [Indexed: 11/17/2022]
Abstract
Microtubules are highly dynamic polymers of α/β tubulin heterodimers that play key roles in cell division and in organizing cell cytoplasm. Although they have been discovered more than two decades ago, tubulin post-translational modifications recently gained a new interest as their role was increasingly highlighted in neuron differentiation and neurodegenerative disorders. Here, we specifically focus on tubulin acetylation from its discovery to recent studies that provide new insights into how it is regulated in health and disease and how it impacts microtubule functions. Even though new mechanisms involving tubulin acetylation are regularly being uncovered, the molecular links between its location inside the microtubule lumen and its regulators and effectors is still poorly understood. This review highlights the emerging roles of tubulin acetylation in multiple cellular functions, ranging from cell motility, cell cycle progression or cell differentiation to intracellular trafficking and signalling. It also points out that tubulin acetylation should no longer be seen as a passive marker of microtubule stability, but as a broad regulator of microtubule functions.
Collapse
Affiliation(s)
- Daniel Perdiz
- Univ. Paris Sud-11, UPRES EA4530 IFR IPSIT, Faculté de Pharmacie, 5 rue JB Clément 92296 Châtenay-Malabry, France
| | | | | | | |
Collapse
|