1
|
Frade AF, Guérin H, Nunes JPS, Silva LFSE, Roda VMDP, Madeira RP, Brochet P, Andrieux P, Kalil J, Chevillard C, Cunha-Neto E. Cardiac and Digestive Forms of Chagas Disease: An Update on Pathogenesis, Genetics, and Therapeutic Targets. Mediators Inflamm 2025; 2025:8862004. [PMID: 40297326 PMCID: PMC12037249 DOI: 10.1155/mi/8862004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 02/12/2025] [Indexed: 04/30/2025] Open
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi (T. cruzi), is a neglected disease affecting around 6 million people, with no effective antiparasitic drugs or vaccines. About 40% of Chagas disease patients develop symptomatic forms in the chronic phase of infection, chronic Chagas cardiomyopathy (CCC) or digestive forms like megaoesophagus and megacolon, while most infected patients (60%) remain asymptomatic (ASY) in the so-called indeterminate form (IF). CCC is an inflammatory cardiomyopathy that occurs decades after the initial infection. Death results from heart failure or arrhythmia in a subset of CCC patients. Myocardial fibrosis, inflammation, and mitochondrial dysfunction are involved in heart failure and arrhythmia. Survival in CCC is worse than in other cardiomyopathies. Distinct from other cardiomyopathies, CCC displays a helper T-cell type 1 (Th1-T) cell-rich myocarditis with abundant interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) and selectively lower levels of mitochondrial energy metabolism enzymes and high-energy phosphates in the heart. A CD8+ T cell-rich inflammatory infiltrate has also been found in the Chagasic megaesophagus, which is associated with denervation of myoenteric plexi. IFN-γ and TNF-α signaling, which are constitutively upregulated in Chagas disease patients, negatively affect mitochondrial function and adenosine 5'-triphosphate (ATP) production-cytokine-induced mitochondrial dysfunction. In addition, the differential susceptibility to developing CCC has prompted many studies over the past 25 years on the association of genetic polymorphisms with disease outcomes. A comprehensive understanding of Chagas disease pathogenesis is crucial for identifying potential therapeutic targets. Genetic studies may offer valuable insights into factors with prognostic significance. In this review, we present an updated perspective on the pathogenesis and genetic factors associated with Chagas disease, emphasizing key studies that elucidate the differential progression of patients to CCC and other symptomatic forms. Furthermore, we explore the interplay between genetic susceptibility, inflammatory cytokines, mitochondrial dysfunction and discuss emerging therapeutic targets.
Collapse
Affiliation(s)
- Amanda Farage Frade
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-900, Brazil
| | - Hélléa Guérin
- French National Institute for Health and Medical Research (INSERM), UMR U1090, TAGC Theories and Approaches of Genomic Complexity, MarMaRa Institute, Aix Marseille University, Marseille 13288, France
| | - Joao Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-900, Brazil
| | - Luiz Felipe Souza e Silva
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-900, Brazil
| | - Vinicius Moraes de Paiva Roda
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-900, Brazil
| | - Rafael Pedro Madeira
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-900, Brazil
| | - Pauline Brochet
- French National Institute for Health and Medical Research (INSERM), UMR U1090, TAGC Theories and Approaches of Genomic Complexity, MarMaRa Institute, Aix Marseille University, Marseille 13288, France
| | - Pauline Andrieux
- French National Institute for Health and Medical Research (INSERM), UMR U1090, TAGC Theories and Approaches of Genomic Complexity, MarMaRa Institute, Aix Marseille University, Marseille 13288, France
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-900, Brazil
- Institute for Investigation in Immunology (III), National Institute of Science and Technology (INCT), São Paulo 05403-900, Brazil
- Department of Clinical Immunology and Allergy, University of São Paulo Medical School, São Paulo 01246-903, Brazil
| | - Christophe Chevillard
- French National Institute for Health and Medical Research (INSERM), UMR U1090, TAGC Theories and Approaches of Genomic Complexity, MarMaRa Institute, Aix Marseille University, Marseille 13288, France
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-900, Brazil
- Institute for Investigation in Immunology (III), National Institute of Science and Technology (INCT), São Paulo 05403-900, Brazil
- Department of Clinical Immunology and Allergy, University of São Paulo Medical School, São Paulo 01246-903, Brazil
| |
Collapse
|
2
|
Michalak KP, Michalak AZ. Understanding chronic inflammation: couplings between cytokines, ROS, NO, Ca i 2+, HIF-1α, Nrf2 and autophagy. Front Immunol 2025; 16:1558263. [PMID: 40264757 PMCID: PMC12012389 DOI: 10.3389/fimmu.2025.1558263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Chronic inflammation is an important component of many diseases, including autoimmune diseases, intracellular infections, dysbiosis and degenerative diseases. An important element of this state is the mainly positive feedback between inflammatory cytokines, reactive oxygen species (ROS), nitric oxide (NO), increased intracellular calcium, hypoxia-inducible factor 1-alpha (HIF-1α) stabilisation and mitochondrial oxidative stress, which, under normal conditions, enhance the response against pathogens. Autophagy and the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response are mainly negatively coupled with the above-mentioned elements to maintain the defence response at a level appropriate to the severity of the infection. The current review is the first attempt to build a multidimensional model of cellular self-regulation of chronic inflammation. It describes the feedbacks involved in the inflammatory response and explains the possible pathways by which inflammation becomes chronic. The multiplicity of positive feedbacks suggests that symptomatic treatment of chronic inflammation should focus on inhibiting multiple positive feedbacks to effectively suppress all dysregulated elements including inflammation, oxidative stress, calcium stress, mito-stress and other metabolic disturbances.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
3
|
Chen M, Zhang S, Huang X, Zhang D, Zhu D, Ouyang C, Li Y. The protective effects and mechanism of myricetin in liver diseases (Review). Mol Med Rep 2025; 31:87. [PMID: 39917997 PMCID: PMC11811602 DOI: 10.3892/mmr.2025.13452] [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: 11/06/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Liver diseases have become one of the significant threats to global health. However, there is a lack of effective targeted therapeutic drugs in this field and the existing drugs used for liver disease treatment usually have side‑effects. Traditional Chinese medicine (TCM) has the distinctive advantages of multi‑target and low side‑effects. As a flavonoid with various pharmacological activities such as anti‑tumour, anti‑oxidant, anti‑inflammatory and anti‑bacterial, the TCM myricetin has been widely used in liver disease research. The present work focuses on the role and molecular mechanism of myricetin in liver diseases such as acute liver injury, fatty liver, liver fibrosis and hepatocellular carcinoma. It is a promising reference for further research and application of myricetin in the treatment of liver diseases.
Collapse
Affiliation(s)
- Mi Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Shengnan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Resources and Environmental Science and Engineering, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xingqiong Huang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dandan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dan Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Changhan Ouyang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yankun Li
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
4
|
Sethi N, Khokhar M, Mathur M, Batra Y, Mohandas A, Tomo S, Rao M, Banerjee M. Therapeutic Potential of Nutraceuticals against Drug-Induced Liver Injury. Semin Liver Dis 2024; 44:430-456. [PMID: 39393795 DOI: 10.1055/s-0044-1791559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Drug-induced liver injury (DILI) continues to be a major concern in clinical practice, thus necessitating a need for novel therapeutic approaches to alleviate its impact on hepatic function. This review investigates the therapeutic potential of nutraceuticals against DILI, focusing on examining the underlying molecular mechanisms and cellular pathways. In preclinical and clinical studies, nutraceuticals, such as silymarin, curcumin, and N-acetylcysteine, have demonstrated remarkable efficacy in attenuating liver injury induced by diverse pharmaceutical agents. The molecular mechanisms underlying these hepatoprotective effects involve modulation of oxidative stress, inflammation, and apoptotic pathways. Furthermore, this review examines cellular routes affected by these nutritional components focusing on their influence on hepatocytes, Kupffer cells, and stellate cells. Key evidence highlights that autophagy modulation as well as unfolded protein response are essential cellular processes through which nutraceuticals exert their cytoprotective functions. In conclusion, nutraceuticals are emerging as promising therapeutic agents for mitigating DILI, by targeting different molecular pathways along with cell processes involved in it concurrently.
Collapse
Affiliation(s)
- Namya Sethi
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mitali Mathur
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Yashi Batra
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Amal Mohandas
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Karnataka, India
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
5
|
Li J, Jiang L, Zhao K, Tang Y, Yuan X, Xu Y. MYELOID-DERIVED TLR4-TRIF SIGNALING PATHWAY MEDIATES OXIDATIVE STRESS IN LPS/D-GALN-INDUCED ACUTE LIVER FAILURE. Shock 2024; 62:582-587. [PMID: 39158930 DOI: 10.1097/shk.0000000000002438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
ABSTRACT Background: Acute liver failure (ALF) is a severe clinical syndrome characterized by massive hepatocyte death in a short time due to viruses, drugs, alcohol, or other factors. Oxidative stress is an important pathogenic mechanism of ALF. LPS-induced internalization of toll-like receptor 4 (TLR4) and the subsequent activation of the toll/IL-1R domain-containing adaptor-inducing IFN-beta (TRIF) signaling pathway widely mediate inflammatory responses in a series of diseases. However, whether the TLR4-TRIF signaling pathway contributes to ALF by mediating oxidative stress processes remains unclear. Methods: An ALF mouse model was induced by lipopolysaccharide (LPS)/D-galactosamine (D-GalN). TLR4-TRIF systemic knockout mice and TLR4 conditional knockout mice were used to determine the role of the TLR4-TRIF signaling pathway in ALF. The effects of TLR4 or TRIF deficiency on oxidative stress were investigated. In addition, we examined the protective role of the clodronate liposomes (macrophage scavengers) and the antioxidant N-acetylcysteine (NAC) in ALF. Results: TLR4 or TRIF deficiency significantly alleviated LPS/D-GalN-induced lethality, hepatic dysfunction, and hepatic pathologic injury, which was dependent on myeloid-derived TLR4. Hence, macrophage clearance exhibits a similar protective effect. Mechanically, TLR4 or TRIF deficiency was observed to inhibit oxidative stress by increasing glutathione, while decreasing malondialdehyde, 8-hydroxy-2-deoxyguanosine, and γ-H2AX. Therefore, the pharmacologic antioxidant NAC exhibited significant hepato-protective effects. Conclusions: Targeting myeloid-derived TLR4-TRIF signaling pathway or antioxidant therapy may be a potential therapeutic direction to treat ALF.
Collapse
Affiliation(s)
| | | | - Kai Zhao
- Department of Hematology and Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | | | | |
Collapse
|
6
|
Nakano R, Chogahara I, Ohira M, Imaoka K, Sato S, Bekki T, Sato K, Imaoka Y, Marlen D, Tanaka Y, Ohdan H. Atherosclerosis Deteriorates Liver Ischemia/Reperfusion Injury Via Interferon Regulatory Factor-1 Overexpression in a Murine Model. Transplant Proc 2024; 56:678-685. [PMID: 38433025 DOI: 10.1016/j.transproceed.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/04/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Abdominal aortic calcification (AAC) is associated with cardiovascular-related mortality, along with an elevated risk of coronary, cerebrovascular, and cardiovascular events. Notably, AAC is strongly associated with poor overall and recurrence free survival posthepatectomy for hepatocellular carcinoma. Despite the acknowledged significance of atherosclerosis in systemic inflammation, its response to ischemia/reperfusion injury (IRI) remains poorly elucidated. In this study, we aimed to clarify the impact of atherosclerosis on the liver immune system using a warm IRI mouse model. METHODS Injury was induced in an atherosclerotic mouse model (ApoE-/-) or C57BL/6J wild-type (WT) mice through 70% clamping for 1 hour and analyzed after 6 hours of reperfusion. RESULTS Elevated serum levels of aspartate and alanine aminotransferase, along with histological assessment, indicated considerable damage in the livers of ApoE-/- mice than that in WT mice. This indicates a substantial contribution of atherosclerosis to IRI. Furthermore, T and natural killer (NK) cells in ApoE-/- mouse livers displayed a more inflammatory phenotype than those in WT mouse livers. Reverse transcription-polymerase chain reaction analysis revealed a significant upregulation of interleukin (IL)-15 and its transcriptional regulator, interferon regulatory factor-1 (IRF-1) in ApoE-/- mouse livers compared with that in WT mouse livers. CONCLUSIONS These findings suggest that in an atherosclerotic mouse model, atherosclerosis can mirror intrahepatic immunity, particularly activating liver NK and T cells through IL-15 production, thereby exacerbating hepatic damage. The upregulation of IL-15 expression is associated with IRF-1 overexpression.
Collapse
Affiliation(s)
- Ryosuke Nakano
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Ichiya Chogahara
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Minami-ku, Hiroshima, Japan.
| | - Kouki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Saki Sato
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Tomoaki Bekki
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Koki Sato
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Yuki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Doskali Marlen
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| |
Collapse
|
7
|
Nozu T, Miyagishi S, Ishioh M, Takakusaki K, Okumura T. Imeglimin prevents visceral hypersensitivity and colonic hyperpermeability in irritable bowel syndrome rat model. J Pharmacol Sci 2023; 153:26-30. [PMID: 37524451 DOI: 10.1016/j.jphs.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/14/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
Visceral hypersensitivity and leaky gut, which are mediated via corticotropin-releasing factor (CRF) and Toll-like receptor 4 are key pathophysiology of irritable bowel syndrome (IBS). Metformin was reported to improve these gastrointestinal (GI) changes. In this study, we attempted to determine the effects of imeglimin, which was synthesized from metformin on GI function in IBS rat models. Imeglimin blocked lipopolysaccharide- or CRF-induced visceral hypersensitivity and colonic hyperpermeability. These effects were prevented by compound C or naloxone. These results suggest that imeglimin may be effective for the treatment of IBS by improved visceral sensation and colonic barrier via AMPK and opioid receptor.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan; Center for Medical Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan.
| | - Saori Miyagishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Masatomo Ishioh
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Kaoru Takakusaki
- Division of Neuroscience, Department of Physiology, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Toshikatsu Okumura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| |
Collapse
|
8
|
Liang Z, Yuan ZH, Wang Y, Du ZH, Guo JJ, Xia LL, Shan Y. New Mechanistic Insight into the Protective Effects of Ganoderma lucidum Polysaccharides Against Palmitic Acid-Induced Cell Damage in Porcine Intestinal Epithelial Cell Line IPEC-J2. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221128103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Ganoderma lucidum ( G. lucidum) is one of the well-known mushrooms in China, which has G. lucidum polysaccharides (GLP) that have been widely studied for various biological activities, such as antioxidant, antitumor, antiinflammatory, antiviral, antidiabetes, and immunomodulatory activities. A signal transducer and activator of transcription (STAT) signaling pathway is related to cell proliferation and apoptosis. The relationship between STAT and intestinal protection of GLP is still unknown. We studied the inhibitors AG490 in the STAT pathway and its downstream molecules to analyze the unique effects in the protection of GLP against palmitic acid (PA)-induced porcine intestinal epithelial cells (IPEC-J2) injury. Compared to PA treatment, GLP + PA obviously decreased Ca2+ concentration, H2O2 production, NF-E2-related factor 2 (Nrf2) nuclear translocation, STAT1 and STAT2 protein levels, and increased nuclear factor kappa-B (NF-κB) nuclear translocation and p-STAT3/STAT3 ratio in IPEC-J2 cells. After inhibition of STAT3 signaling, p-STAT3/STAT3 ratio, NF-κB nuclear translocation obviously decreased and Nrf2 nuclear translocation significantly increased in the GLP + PA group. The protection of GLP on proliferation and apoptosis of PA-induced IPEC-J2 cells was suppressed by inhibiting STAT3. The STAT3 pathway regulated the enterocyte-protective effects of GLP by modulating the nuclear translocation of Nrf2 and NF-κB. We provide new insights into the mechanism of STAT signaling for the protection of GLP on PA-induced intestinal epithelial cell injury.
Collapse
Affiliation(s)
- Zengenni Liang
- Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, China
| | | | - Yan Wang
- Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Zhong-Hua Du
- Changsha Qiantu Biological Technology Limited Company, Changsha, China
| | - Jia-Jing Guo
- Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, China
| | - Ling-Li Xia
- Changsha Diwei Agricultural Technology Limited Company, Changsha, China
| | - Yang Shan
- Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, China
| |
Collapse
|
9
|
Silva KDA, Nunes JPS, Andrieux P, Brochet P, Almeida RR, Kuramoto Takara ACK, Pereira NB, Abel L, Cobat A, Zaniratto RCF, Levy D, Bydlowski SP, Cecconello I, Seguro FCBDC, Kalil J, Chevillard C, Cunha-Neto E. Chagas Disease Megaesophagus Patients Carrying Variant MRPS18B P260A Display Nitro-Oxidative Stress and Mitochondrial Dysfunction in Response to IFN-γ Stimulus. Biomedicines 2022; 10:2215. [PMID: 36140315 PMCID: PMC9496350 DOI: 10.3390/biomedicines10092215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas disease (CD), caused by the protozoan parasite Trypanosoma cruzi, affects 8 million people, and around 1/3 develop chronic cardiac (CCC) or digestive disease (megaesophagus/megacolon), while the majority remain asymptomatic, in the indeterminate form of Chagas disease (ASY). Most CCC cases in families with multiple Chagas disease patients carry damaging mutations in mitochondrial genes. We searched for exonic mutations associated to chagasic megaesophagus (CME) in genes essential to mitochondrial processes. We performed whole exome sequencing of 13 CME and 45 ASY patients. We found the damaging variant MRPS18B 688C > G P230A, in five out of the 13 CME patients (one of them being homozygous; 38.4%), while the variant appeared in one out of 45 ASY patients (2.2%). We analyzed the interferon (IFN)-γ-induced nitro-oxidative stress and mitochondrial function of EBV-transformed lymphoblastoid cell lines. We found the CME carriers of the mutation displayed increased levels of nitrite and nitrated proteins; in addition, the homozygous (G/G) CME patient also showed increased mitochondrial superoxide and reduced levels of ATP production. The results suggest that pathogenic mitochondrial mutations may contribute to cytokine-induced nitro-oxidative stress and mitochondrial dysfunction. We hypothesize that, in mutation carriers, IFN-γ produced in the esophageal myenteric plexus might cause nitro-oxidative stress and mitochondrial dysfunction in neurons, contributing to megaesophagus.
Collapse
Affiliation(s)
- Karla Deysiree Alcântara Silva
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - João Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Pauline Andrieux
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Pauline Brochet
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Rafael Ribeiro Almeida
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - Andréia Cristina Kazue Kuramoto Takara
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - Natalia Bueno Pereira
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, University of Paris, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Aurelie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, University of Paris, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Ricardo Costa Fernandes Zaniratto
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Débora Levy
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Ivan Cecconello
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - Christophe Chevillard
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| |
Collapse
|
10
|
Chen CL, Tseng PC, Satria RD, Nguyen TT, Tsai CC, Lin CF. Role of Glycogen Synthase Kinase-3 in Interferon-γ-Mediated Immune Hepatitis. Int J Mol Sci 2022; 23:ijms23094669. [PMID: 35563060 PMCID: PMC9101719 DOI: 10.3390/ijms23094669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3), a serine/threonine kinase, is a vital glycogen synthase regulator controlling glycogen synthesis, glucose metabolism, and insulin signaling. GSK-3 is widely expressed in different types of cells, and its abundant roles in cellular bioregulation have been speculated. Abnormal GSK-3 activation and inactivation may affect its original bioactivity. Moreover, active and inactive GSK-3 can regulate several cytosolic factors and modulate their diverse cellular functional roles. Studies in experimental liver disease models have illustrated the possible pathological role of GSK-3 in facilitating acute hepatic injury. Pharmacologically targeting GSK-3 is therefore suggested as a therapeutic strategy for liver protection. Furthermore, while the signaling transduction of GSK-3 facilitates proinflammatory interferon (IFN)-γ in vitro and in vivo, the blockade of GSK-3 can be protective, as shown by an IFN-γ-induced immune hepatitis model. In this study, we explored the possible regulation of GSK-3 and the potential relevance of GSK-3 blockade in IFN-γ-mediated immune hepatitis.
Collapse
Affiliation(s)
- Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Po-Chun Tseng
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan;
| | - Rahmat Dani Satria
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (R.D.S.); (T.T.N.)
- Department of Clinical Pathology and Laboratory Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Clinical Laboratory Installation, Dr. Sardjito Central General Hospital, Yogyakarta 55281, Indonesia
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Thi Thuy Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (R.D.S.); (T.T.N.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Oncology, Hue University of Medicine and Pharmacy, Hue University, Hue City 530000, Vietnam
| | - Cheng-Chieh Tsai
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan 703, Taiwan
- Department of Long Term Care Management, Chung Hwa University of Medical Technology, Tainan 703, Taiwan
- Correspondence: (C.-C.T.); (C.-F.L.)
| | - Chiou-Feng Lin
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan;
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (R.D.S.); (T.T.N.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (C.-C.T.); (C.-F.L.)
| |
Collapse
|
11
|
Teixeira PC, Ducret A, Langen H, Nogoceke E, Santos RHB, Silva Nunes JP, Benvenuti L, Levy D, Bydlowski SP, Bocchi EA, Kuramoto Takara A, Fiorelli AI, Stolf NA, Pomeranzeff P, Chevillard C, Kalil J, Cunha-Neto E. Impairment of Multiple Mitochondrial Energy Metabolism Pathways in the Heart of Chagas Disease Cardiomyopathy Patients. Front Immunol 2021; 12:755782. [PMID: 34867990 PMCID: PMC8633876 DOI: 10.3389/fimmu.2021.755782] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
Chagas disease cardiomyopathy (CCC) is an inflammatory dilated cardiomyopathy occurring in 30% of the 6 million infected with the protozoan Trypanosoma cruzi in Latin America. Survival is significantly lower in CCC than ischemic (IC) and idiopathic dilated cardiomyopathy (DCM). Previous studies disclosed a selective decrease in mitochondrial ATP synthase alpha expression and creatine kinase activity in CCC myocardium as compared to IDC and IC, as well as decreased in vivo myocardial ATP production. Aiming to identify additional constraints in energy metabolism specific to CCC, we performed a proteomic study in myocardial tissue samples from CCC, IC and DCM obtained at transplantation, in comparison with control myocardial tissue samples from organ donors. Left ventricle free wall myocardial samples were subject to two-dimensional electrophoresis with fluorescent labeling (2D-DIGE) and protein identification by mass spectrometry. We found altered expression of proteins related to mitochondrial energy metabolism, cardiac remodeling, and oxidative stress in the 3 patient groups. Pathways analysis of proteins differentially expressed in CCC disclosed mitochondrial dysfunction, fatty acid metabolism and transmembrane potential of mitochondria. CCC patients’ myocardium displayed reduced expression of 22 mitochondrial proteins belonging to energy metabolism pathways, as compared to 17 in DCM and 3 in IC. Significantly, 6 beta-oxidation enzymes were reduced in CCC, while only 2 of them were down-regulated in DCM and 1 in IC. We also observed that the cytokine IFN-gamma, previously described with increased levels in CCC, reduces mitochondrial membrane potential in cardiomyocytes. Results suggest a major reduction of mitochondrial energy metabolism and mitochondrial dysfunction in CCC myocardium which may be in part linked to IFN-gamma. This may partially explain the worse prognosis of CCC as compared to DCM or IC.
Collapse
Affiliation(s)
- Priscila Camillo Teixeira
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Axel Ducret
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Hanno Langen
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Everson Nogoceke
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | | | - João Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto Nacional de Ciência e Tecnologia, INCT, iii- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Luiz Benvenuti
- Anatomical Pathology Division, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Debora Levy
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Edimar Alcides Bocchi
- Heart Failure Team, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Andréia Kuramoto Takara
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Alfredo Inácio Fiorelli
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Noedir Antonio Stolf
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pablo Pomeranzeff
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Christophe Chevillard
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto Nacional de Ciência e Tecnologia, INCT, iii- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto Nacional de Ciência e Tecnologia, INCT, iii- Institute for Investigation in Immunology, São Paulo, Brazil
| |
Collapse
|
12
|
Nunes JPS, Andrieux P, Brochet P, Almeida RR, Kitano E, Honda AK, Iwai LK, Andrade-Silva D, Goudenège D, Alcântara Silva KD, Vieira RDS, Levy D, Bydlowski SP, Gallardo F, Torres M, Bocchi EA, Mano M, Santos RHB, Bacal F, Pomerantzeff P, Laurindo FRM, Teixeira PC, Nakaya HI, Kalil J, Procaccio V, Chevillard C, Cunha-Neto E. Co-Exposure of Cardiomyocytes to IFN-γ and TNF-α Induces Mitochondrial Dysfunction and Nitro-Oxidative Stress: Implications for the Pathogenesis of Chronic Chagas Disease Cardiomyopathy. Front Immunol 2021; 12:755862. [PMID: 34867992 PMCID: PMC8632642 DOI: 10.3389/fimmu.2021.755862] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
Infection by the protozoan Trypanosoma cruzi causes Chagas disease cardiomyopathy (CCC) and can lead to arrhythmia, heart failure and death. Chagas disease affects 8 million people worldwide, and chronic production of the cytokines IFN-γ and TNF-α by T cells together with mitochondrial dysfunction are important players for the poor prognosis of the disease. Mitochondria occupy 40% of the cardiomyocytes volume and produce 95% of cellular ATP that sustain the life-long cycles of heart contraction. As IFN-γ and TNF-α have been described to affect mitochondrial function, we hypothesized that IFN-γ and TNF-α are involved in the myocardial mitochondrial dysfunction observed in CCC patients. In this study, we quantified markers of mitochondrial dysfunction and nitro-oxidative stress in CCC heart tissue and in IFN-γ/TNF-α-stimulated AC-16 human cardiomyocytes. We found that CCC myocardium displayed increased levels of nitro-oxidative stress and reduced mitochondrial DNA as compared with myocardial tissue from patients with dilated cardiomyopathy (DCM). IFN-γ/TNF-α treatment of AC-16 cardiomyocytes induced increased nitro-oxidative stress and decreased the mitochondrial membrane potential (ΔΨm). We found that the STAT1/NF-κB/NOS2 axis is involved in the IFN-γ/TNF-α-induced decrease of ΔΨm in AC-16 cardiomyocytes. Furthermore, treatment with mitochondria-sparing agonists of AMPK, NRF2 and SIRT1 rescues ΔΨm in IFN-γ/TNF-α-stimulated cells. Proteomic and gene expression analyses revealed that IFN-γ/TNF-α-treated cells corroborate mitochondrial dysfunction, transmembrane potential of mitochondria, altered fatty acid metabolism and cardiac necrosis/cell death. Functional assays conducted on Seahorse respirometer showed that cytokine-stimulated cells display decreased glycolytic and mitochondrial ATP production, dependency of fatty acid oxidation as well as increased proton leak and non-mitochondrial oxygen consumption. Together, our results suggest that IFN-γ and TNF-α cause direct damage to cardiomyocytes’ mitochondria by promoting oxidative and nitrosative stress and impairing energy production pathways. We hypothesize that treatment with agonists of AMPK, NRF2 and SIRT1 might be an approach to ameliorate the progression of Chagas disease cardiomyopathy.
Collapse
Affiliation(s)
- João Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil.,INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Pauline Andrieux
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Pauline Brochet
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Rafael Ribeiro Almeida
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Eduardo Kitano
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - André Kenji Honda
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Leo Kei Iwai
- Laboratório Especial de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Débora Andrade-Silva
- Laboratório Especial de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - David Goudenège
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
| | - Karla Deysiree Alcântara Silva
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Raquel de Souza Vieira
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Débora Levy
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Frédéric Gallardo
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Magali Torres
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Edimar Alcides Bocchi
- Heart Failure Team, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | | | - Fernando Bacal
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pablo Pomerantzeff
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Priscila Camillo Teixeira
- Translational Research Sciences, Pharma Research and Early Development F. Hoffmann-La Roche, Basel, Switzerland
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Vincent Procaccio
- MitoLab, UMR CNRS 6015-INSERM U1083, Université d'Angers, Angers, France
| | - Christophe Chevillard
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| |
Collapse
|
13
|
Woznicki JA, Saini N, Flood P, Rajaram S, Lee CM, Stamou P, Skowyra A, Bustamante-Garrido M, Regazzoni K, Crawford N, McDade SS, Longley DB, Aza-Blanc P, Shanahan F, Zulquernain SA, McCarthy J, Melgar S, McRae BL, Nally K. TNF-α synergises with IFN-γ to induce caspase-8-JAK1/2-STAT1-dependent death of intestinal epithelial cells. Cell Death Dis 2021; 12:864. [PMID: 34556638 PMCID: PMC8459343 DOI: 10.1038/s41419-021-04151-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/16/2021] [Accepted: 09/08/2021] [Indexed: 12/25/2022]
Abstract
Rewiring of host cytokine networks is a key feature of inflammatory bowel diseases (IBD) such as Crohn's disease (CD). Th1-type cytokines-IFN-γ and TNF-α-occupy critical nodes within these networks and both are associated with disruption of gut epithelial barrier function. This may be due to their ability to synergistically trigger the death of intestinal epithelial cells (IECs) via largely unknown mechanisms. In this study, through unbiased kinome RNAi and drug repurposing screens we identified JAK1/2 kinases as the principal and nonredundant drivers of the synergistic killing of human IECs by IFN-γ/TNF-α. Sensitivity to IFN-γ/TNF-α-mediated synergistic IEC death was retained in primary patient-derived intestinal organoids. Dependence on JAK1/2 was confirmed using genetic loss-of-function studies and JAK inhibitors (JAKinibs). Despite the presence of biochemical features consistent with canonical TNFR1-mediated apoptosis and necroptosis, IFN-γ/TNF-α-induced IEC death was independent of RIPK1/3, ZBP1, MLKL or caspase activity. Instead, it involved sustained activation of JAK1/2-STAT1 signalling, which required a nonenzymatic scaffold function of caspase-8 (CASP8). Further modelling in gut mucosal biopsies revealed an intercorrelated induction of the lethal CASP8-JAK1/2-STAT1 module during ex vivo stimulation of T cells. Functional studies in CD-derived organoids using inhibitors of apoptosis, necroptosis and JAKinibs confirmed the causative role of JAK1/2-STAT1 in cytokine-induced death of primary IECs. Collectively, we demonstrate that TNF-α synergises with IFN-γ to kill IECs via the CASP8-JAK1/2-STAT1 module independently of canonical TNFR1 and cell death signalling. This non-canonical cell death pathway may underpin immunopathology driven by IFN-γ/TNF-α in diverse autoinflammatory diseases such as IBD, and its inhibition may contribute to the therapeutic efficacy of anti-TNFs and JAKinibs.
Collapse
Affiliation(s)
| | - Nisha Saini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Peter Flood
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Ciaran M Lee
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | | | | | | | - Nyree Crawford
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Simon S McDade
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Daniel B Longley
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Pedro Aza-Blanc
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland
| | - Syed A Zulquernain
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland
| | - Jane McCarthy
- Department of Gastroenterology, Mercy University Hospital, Cork, Ireland
| | - Silvia Melgar
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Bradford L McRae
- Immunology Discovery, Abbvie Bioresearch Center, Worcester, MA, 01605, USA
| | - Ken Nally
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| |
Collapse
|
14
|
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are clinical syndromes that cause significant mortality in clinical settings and morbidity among survivors accompanied by huge healthcare costs. Lung-resident cell dysfunction/death and neutrophil alveolitis accompanied by proteinous edema are the main pathological features of ALI/ARDS. While understanding of the mechanisms underlying ALI/ARDS pathogenesis is progressing and potential treatments such as statin therapy, nutritional strategies, and mesenchymal cell therapy are emerging, poor clinical outcomes in ALI/ARDS patients persist. Thus, a better understanding of lung-resident cell death and neutrophil alveolitis and their mitigation and clearance mechanisms may provide new therapeutic strategies to accelerate lung repair and improve outcomes in critically ill patients. Macrophages are required for normal tissue development and homeostasis as well as regulating tissue injury and repair through modulation of inflammation and other cellular processes. While macrophages mediate various functions, here we review recent dead cell clearance (efferocytosis) mechanisms mediated by these immune cells for maintaining tissue homeostasis after infectious and non-infectious lung injury.
Collapse
Affiliation(s)
- Patrick M Noone
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | - Sekhar P Reddy
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, IL 60612, USA
- Department of Pathology, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| |
Collapse
|
15
|
Rare Pathogenic Variants in Mitochondrial and Inflammation-Associated Genes May Lead to Inflammatory Cardiomyopathy in Chagas Disease. J Clin Immunol 2021; 41:1048-1063. [PMID: 33660144 PMCID: PMC8249271 DOI: 10.1007/s10875-021-01000-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/15/2021] [Indexed: 01/21/2023]
Abstract
Abstract Cardiomyopathies are an important cause of heart failure and sudden cardiac death. Little is known about the role of rare genetic variants in inflammatory cardiomyopathy. Chronic Chagas disease cardiomyopathy (CCC) is an inflammatory cardiomyopathy prevalent in Latin America, developing in 30% of the 6 million patients chronically infected by the protozoan Trypanosoma cruzi, while 60% remain free of heart disease (asymptomatic (ASY)). The cytokine interferon-γ and mitochondrial dysfunction are known to play a major pathogenetic role. Chagas disease provides a unique model to probe for genetic variants involved in inflammatory cardiomyopathy. Methods We used whole exome sequencing to study nuclear families containing multiple cases of Chagas disease. We searched for rare pathogenic variants shared by all family members with CCC but absent in infected ASY siblings and in unrelated ASY. Results We identified heterozygous, pathogenic variants linked to CCC in all tested families on 22 distinct genes, from which 20 were mitochondrial or inflammation-related – most of the latter involved in proinflammatory cytokine production. Significantly, incubation with IFN-γ on a human cardiomyocyte line treated with an inhibitor of dihydroorotate dehydrogenase brequinar (enzyme showing a loss-of-function variant in one family) markedly reduced mitochondrial membrane potential (ΔψM), indicating mitochondrial dysfunction. Conclusion Mitochondrial dysfunction and inflammation may be genetically determined in CCC, driven by rare genetic variants. We hypothesize that CCC-linked genetic variants increase mitochondrial susceptibility to IFN-γ-induced damage in the myocardium, leading to the cardiomyopathy phenotype in Chagas disease. This mechanism may also be operative in other inflammatory cardiomyopathies. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-021-01000-y.
Collapse
|
16
|
Wei W, Xiao X, Li J, Ding H, Pan W, Deng S, Yin W, Xue L, Lu Q, Yue Y, Tian Y, Wang M, Hao L. Activation of the STAT1 Pathway Accelerates Periodontitis in Nos3-/- Mice. J Dent Res 2020; 98:1027-1036. [PMID: 31329047 DOI: 10.1177/0022034519858063] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Early studies on the etiology and pathogenesis of hypertension have shown that it has a considerable association with inflammation and the immune response as well as periodontitis. Clinical studies have also shown that hypertension can promote the periodontal tissue destruction caused by periodontitis. However, the underlying mechanisms remain unclear. This study aimed to explore the possible mechanisms of how hypertension aggravates periodontitis. Treatment with or without the signal transducer and activator of transcription 1 (STAT1) inhibitor fludarabine was performed in an endothelial nitric oxide synthase gene knockout-related (Nos3-/-) mouse model with the hypertension phenotype of periodontitis induced by bacteria. Micro-computed tomography, immunohistochemistry, Western blot, quantitative reverse transcription polymerase chain reaction, immunofluorescence, and ELISA were performed. We demonstrated that Nos3-/--related hypertension increases bone resorption and periodontal destruction in periodontitis lesion areas, which can be inhibited by the STAT1 inhibitor. Experimental data also showed that Nos3-/- significantly increased macrophage infiltration and proinflammatory cytokine expression in the periodontitis lesion area, which is dependent on the angiotensin II-induced STAT1 pathway. Inhibition of STAT1 in vivo can decrease the expression of proinflammatory cytokines and macrophage infiltration. Furthermore, data in this study showed that Nos3-/--related hypertension further downregulated the STAT3 anti-inflammatory function and its downstream chemokine expression in a STAT1-dependent manner. By applying RAW 264.7 and L929 cell lines and monocytes isolated from Nos3-/- mice, we confirmed that activation of the STAT1 pathway inhibits STAT3 and its downstream pathway and promotes inflammatory cytokine expression in vitro. Collectively, our current study demonstrated that STAT1 plays an indispensable role in the Nos3-/--related hypertension with aggravation of periodontitis, suggesting that STAT1 may be a key target for the treatment of periodontitis with hypertension.
Collapse
Affiliation(s)
- W Wei
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - X Xiao
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - J Li
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - H Ding
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - W Pan
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - S Deng
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - W Yin
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - L Xue
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Q Lu
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Y Yue
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Y Tian
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - M Wang
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - L Hao
- 1 The State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| |
Collapse
|
17
|
Pibiri M. Liver regeneration in aged mice: new insights. Aging (Albany NY) 2019; 10:1801-1824. [PMID: 30157472 PMCID: PMC6128415 DOI: 10.18632/aging.101524] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023]
Abstract
The regenerative capacity of the liver after resection is reduced with aging. Recent studies on rodents revealed that both intracellular and extracellular factors are involved in the impairment of liver mass recovery during aging. Among the intracellular factors, age-dependent decrease of BubR1 (budding uninhibited by benzimidazole-related 1), YAP (Yes-associated protein) and SIRT1 (Sirtuin-1) have been associated to dampening of tissue reconstitution and inhibition of cell cycle genes following partial hepatectomy. Extra-cellular factors, such as age-dependent changes in hepatic stellate cells affect liver regeneration through inhibition of progenitor cells and reduction of liver perfusion. Furthermore, chronic release of pro-inflammatory proteins by senescent cells (SASP) affects cell proliferation suggesting that senescent cell clearance might improve tissue regeneration. Accordingly, young plasma restores liver regeneration in aged animals through autophagy re-establishment. This review will discuss how intracellular and extracellular factors cooperate to guarantee a proper liver regeneration and the possible causes of its impairment during aging. The possibility that an improvement of the liver regenerative capacity in elderly might be achieved through elimination of senescent cells via autophagy or by administration of direct mitogenic agents devoid of cytotoxicity will also be entertained.
Collapse
Affiliation(s)
- Monica Pibiri
- Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Cagliari 09124, Italy
| |
Collapse
|
18
|
IRF-1 Intervention in the Classical ROS-Dependent Release of NETs during LPS-Induced Acute Lung Injury in Mice. Inflammation 2019; 42:387-403. [PMID: 30315525 DOI: 10.1007/s10753-018-0903-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previously, we demonstrated that neutrophil extracellular traps (NETs) play an essential role in lipopolysaccharide (LPS)-induced acute lung injury. However, the underlying mechanism is unclear. In this study, we showed that knockout of interferon regulatory factor 1 (IRF-1) in mice strongly attenuated the generation of NETs and reactive oxygen species (ROS) production in neutrophils from bronchoalveolar lavage fluid and alleviated LPS-induced lung injury and systemic inflammation. Our in vitro experiments demonstrated that LPS-stimulated platelets induce NET release through two distinct processes: an ROS-independent early/rapid NETosis and a later ROS-dependent classical NETosis. Notably, the classical ROS-dependent pathway plays a dominant role in the generation of NETs. Furthermore, we showed that IRF-1 knockout does not affect the formation of NETs in early/rapid NETosis, but significantly attenuates ROS production and the generation of NETs in classical NETosis, which determines the total levels of NETs released by LPS-stimulated platelets. In conclusion, IRF-1 deficiency plays a key role in moderating the excessive NETs formed via ROS in the classical pathway and retaining the protective role of the low-NET levels generated in early/rapid NETosis, which may serve as a novel target in acute lung injury/acute respiratory distress syndrome.
Collapse
|
19
|
Bai K, Jiang L, Zhu S, Feng C, Zhao Y, Zhang L, Wang T. Dimethylglycine sodium salt protects against oxidative damage and mitochondrial dysfunction in the small intestines of mice. Int J Mol Med 2019; 43:2199-2211. [PMID: 30816456 DOI: 10.3892/ijmm.2019.4093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 02/07/2019] [Indexed: 11/06/2022] Open
Abstract
Few studies have investigated the use of dimethylglycine sodium salt (DMG‑Na) to protect against small intestinal damage, despite its prevalence in the treatment of human diseases. The present study aimed to evaluate the protective effects of DMG‑Na against oxidative damage and mitochondrial dysfunction in the small intestines of mice. A total of 100 male Kunming mice were randomly assigned to five groups (n=20 per group): i) Mice gastric intubation with 0.3 ml sterile saline solution (once), then subcutaneously injected with sterile saline solution (0.5 ml) after 1 h (CON); ii) mice gastric intubation with 12 mg DMG‑Na/0.3 ml of sterile saline solution once, then subcutaneously injected with sterile saline solution (0.5 ml) 1 h later (D); iii) mice gastric intubation with 0.3 ml sterile saline solution once, then subcutaneously injected with indomethacin (10 mg/kg BW) 1 h later (IN); iv) mice gastric intubation with 12 mg DMG‑Na/0.3 ml sterile saline solution once, then subcutaneously injected with indomethacin (10 mg/kg BW) 1 h later (DIN); and v) mice subcutaneously injected with indomethacin (10 mg/kg BW), then gastrically intubated with 12 mg DMG‑Na/0.3 ml sterile saline solution once after 1 h (IND). The present study was evaluated the effects of DMG‑Na on mice intestinal damage induced by indomethacin injection. The histological morphology of the small intestine improved (P<0.05) in the DIN and IND groups, compared with the IN group. The antioxidant system was enhanced, oxidative damage was reduced, and the expression of antioxidant‑associated genes was increased in the small intestine and its mitochondria in the DIN and IND groups, compared with the IN group. The above results suggested that pretreatment and treatment with DMG‑Na reduced oxidative damage by enhancing antioxidant capacity, increasing the expression of antioxidant‑associated genes, ameliorating mitochondrial dysfunction and suppressing apoptosis. Further study is required to determine the specific mechanism by which pretreatment and treatment with DMG‑Na reduced small intestinal damage.
Collapse
Affiliation(s)
- Kaiwen Bai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Luyi Jiang
- College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Shanli Zhu
- College of Agriculture and Life Science, Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
| | - Chengcheng Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yongwei Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| |
Collapse
|
20
|
Chevillard C, Nunes JPS, Frade AF, Almeida RR, Pandey RP, Nascimento MS, Kalil J, Cunha-Neto E. Disease Tolerance and Pathogen Resistance Genes May Underlie Trypanosoma cruzi Persistence and Differential Progression to Chagas Disease Cardiomyopathy. Front Immunol 2018; 9:2791. [PMID: 30559742 PMCID: PMC6286977 DOI: 10.3389/fimmu.2018.02791] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 11/13/2018] [Indexed: 01/01/2023] Open
Abstract
Chagas disease is caused by infection with the protozoan Trypanosoma cruzi and affects over 8 million people worldwide. In spite of a powerful innate and adaptive immune response in acute infection, the parasite evades eradication, leading to a chronic persistent infection with low parasitism. Chronically infected subjects display differential patterns of disease progression. While 30% develop chronic Chagas disease cardiomyopathy (CCC)-a severe inflammatory dilated cardiomyopathy-decades after infection, 60% of the patients remain disease-free, in the asymptomatic/indeterminate (ASY) form, and 10% develop gastrointestinal disease. Infection of genetically deficient mice provided a map of genes relevant for resistance to T. cruzi infection, leading to the identification of multiple genes linked to survival to infection. These include pathogen resistance genes (PRG) needed for intracellular parasite destruction, and genes involved in disease tolerance (protection against tissue damage and acute phase death-DTG). All identified DTGs were found to directly or indirectly inhibit IFN-γ production or Th1 differentiation. We hypothesize that the absolute need for DTG to control potentially lethal IFN-γ PRG activity leads to T. cruzi persistence and establishment of chronic infection. IFN-γ production is higher in CCC than ASY patients, and is the most highly expressed cytokine in CCC hearts. Key DTGs that downmodulate IFN-γ, like IL-10, and Ebi3/IL27p28, are higher in ASY patients. Polymorphisms in PRG and DTG are associated with differential disease progression. We thus hypothesize that ASY patients are disease tolerant, while an imbalance of DTG and IFN-γ PRG activity leads to the inflammatory heart damage of CCC.
Collapse
Affiliation(s)
| | - João Paulo Silva Nunes
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Amanda Farage Frade
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
- Department of Bioengineering, Brazil University, São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Ramendra Pati Pandey
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Marilda Savóia Nascimento
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Jorge Kalil
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| |
Collapse
|
21
|
Bai K, Jiang L, Zhang L, Zhao Y, Lu Y, Zhu J, Cai J, Zhang L, Wang T. In vitro free radical scavenging capacity of dimethylglycine sodium salt and its protective ability against oleic acid hydroperoxide-induced oxidative damage in IPEC-J2 cells. Int J Mol Med 2018; 42:3447-3458. [PMID: 30221672 DOI: 10.3892/ijmm.2018.3876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/14/2018] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to evaluate the in vitro free radical scavenging capacity of dimethylglycine sodium (DMG‑Na) and its protective ability against oleic acid hydroperoxide (OAHPx)‑induced oxidative damage in IPEC‑J2 cells. Initially, the free radical scavenging activities of water‑soluble pigments (DMG‑Na, betalain, capsanthin and cyanidin‑3‑rutinoside) were measured and compared with those of Trolox. Subsequently, freshly collected swine blood was mixed with heparin and centrifuged to obtain erythrocytes. In order to induce the free radical chain oxidation in erythrocytes, the aqueous peroxyl radicals were generated by thermal decomposition of 2,2'‑azobis(2‑amidinopropane) dihydrochloride (AAPH) in oxygen. A 2% suspension of porcine erythrocytes in PBS buffer were pre‑incubated for 30 min at 37˚C with DMG‑Na (32 µM), followed by incubation with or without AAPH (75 mM) for 5 h with gentle shaking. Additionally, IPEC‑J2 cells were randomly assigned to four groups (n=6 per group): Cells treated with phosphate buffered saline (PBS); cells treated with DMG‑Na (32 µM); cells treated with oleic acid hydroperoxides (OAHPx, 20 µM; TO group); cells treated with DMG‑Na (32 µM) followed by OAHPx (20 µM; DTO group). The cells were cultured in Dulbecco's modified Eagle's medium, Ham's F‑12 mixture, 1.5 mM HEPES, 5% (v/v) fetal bovine serum, 1% (v/v) insulin‑transferrin‑selenium mixture, 1% (v/v) penicillin‑streptomycin mixture and 2.5 µg/ml fungizone (37˚C, 5% CO2). The results showed that DMG‑Na exerted the strongest free radical scavenging capacity at 0.32 M from 0.08‑0.64 M, and that it could prevent AAPH‑induced porcine erythrocyte hemolysis by increasing its antioxidant capacity (P<0.05). The results also demonstrated that antioxidant capacity and antioxidant‑associated gene expression increased in the DTO group relative to the TO group (P<0.05), indicating that DMG‑Na prevented the OAHPx‑induced oxidative damage in IPEC‑J2 cells by improving the antioxidant capacity and antioxidant‑associated gene expression.
Collapse
Affiliation(s)
- Kaiwen Bai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Luyi Jiang
- College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Ligen Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yongwei Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yi Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Jingya Zhu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Jie Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| |
Collapse
|
22
|
Bak DH, Na J, Choi MJ, Lee BC, Oh CT, Kim JY, Han HJ, Kim MJ, Kim TH, Kim BJ. Anti‑apoptotic effects of human placental hydrolysate against hepatocyte toxicity in vivo and in vitro. Int J Mol Med 2018; 42:2569-2583. [PMID: 30132515 PMCID: PMC6192762 DOI: 10.3892/ijmm.2018.3830] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023] Open
Abstract
Apoptosis and oxidative stress are essential for the pathogenesis of acute liver failure and fulminant hepatic failure. Human placental hydrolysate (hPH) has been reported to possess antioxidant and anti-inflammatory properties. In the present study, the protective effects of hPH against D-galactosamine (D-GalN)- and lipopolysaccharide (LPS)-induced hepatocyte apoptosis were investigated in vivo. In addition, the molecular mechanisms underlying the anti-apoptotic activities of hPH against D-GalN-induced cell death in vitro were examined. Male Sprague-Dawley rats were injected with D-GaIN/LPS with or without the administration of hPH. Rats were sacrificed 24 h after D-GaIN/LPS intraperitoneal injection, and the blood and liver samples were collected for future inflammation and hepatotoxicity analyses. Changes in cell viability, apoptosis protein expression, mitochondrial mass, mitochondrial membrane potential, reactive oxygen species generation, and the levels of proteins and mRNA associated with a protective mechanism were determined in HepG2 cells pretreated with hPH for 2 h prior to D-GalN exposure. The findings suggested that hPH treatment effectively protected against D-GalN/LPS-induced hepatocyte apoptosis by reducing the levels of alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase, interleukin-6, and tumor necrosis factor-α, and increasing the level of proliferating cell nuclear antigen. It was also found that hPH inhibited the apoptotic cell death induced by D-GalN. hPH activated the expression of antioxidant enzymes, including superoxide dismutase, glutathione peroxidase, and catalase, which were further upregulated by the Kelch-like ECH2-associated protein 1-p62-nuclear factor-erythroid 2-related factor 2 pathway, a component of oxidative stress defense mechanisms. Furthermore, hPH markedly reduced cytosolic and mitochondrial reactive oxygen species and rescued mitochondrial loss and dysfunction through the reduction of damage-regulated autophagy modulator, p53, and C/EBP homologous protein. Collectively, hPH exhibited a protective role in hepatocyte apoptosis by inhibiting oxidative stress and maintaining cell homeostasis. The underlying mechanisms may be associated with the inhibition of endoplasmic reticulum stress and minimization of the autophagy progress.
Collapse
Affiliation(s)
- Dong-Ho Bak
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06973, Republic of Korea
| | - Jungtae Na
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06973, Republic of Korea
| | - Mi Ji Choi
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06973, Republic of Korea
| | - Byung Chul Lee
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06973, Republic of Korea
| | - Chang Taek Oh
- Research and Development Center, Green Cross WellBeing Corporation, Seongnam, Gyeonggi 13595, Republic of Korea
| | - Jeom-Yong Kim
- Research and Development Center, Green Cross WellBeing Corporation, Seongnam, Gyeonggi 13595, Republic of Korea
| | - Hae Jung Han
- Research and Development Center, Green Cross WellBeing Corporation, Seongnam, Gyeonggi 13595, Republic of Korea
| | | | - Tae Ho Kim
- Division of Gastroenterology, Department of Internal Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, Bucheon‑si, Gyeonggi 14647, Republic of Korea
| | - Beom Joon Kim
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06973, Republic of Korea
| |
Collapse
|
23
|
Cao S, Zhang Q, Wang C, Wu H, Jiao L, Hong Q, Hu C. LPS challenge increased intestinal permeability, disrupted mitochondrial function and triggered mitophagy of piglets. Innate Immun 2018; 24:221-230. [PMID: 29642727 PMCID: PMC6830921 DOI: 10.1177/1753425918769372] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/07/2018] [Accepted: 03/15/2018] [Indexed: 12/21/2022] Open
Abstract
Here we investigated the influence of LPS-induced gut injury on antioxidant homeostasis, mitochondrial (mt) function and the level of mitophagy in piglets. The results showed that LPS-induced intestinal injury decreased the transepithelial electrical resistance, increased the paracellular permeability of F1TC dextran 4 kDa, and decreased the expression of claudin-1, occludin and zonula occludens-1 in the jejunum compared with the control group. LPS decreased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and increased the content of malondialdehyde in the jejunum. Meanwhile, the expression of SOD-related genes ( Cu/Zn-SOD, Mn-SOD) and GSH-Px-related genes ( GPX-1, GPX-4) declined in LPS-challenged pigs compared with the control. LPS also increased TNF-α, IL-6, IL-8 and IL-1β mRNA expression. LPS induced mt dysfunction, as demonstrated by increased reactive oxygen species production and decreased membrane potential of intestinal mitochondria, intestinal content of mt DNA and activities of the intestinal mt respiratory chain. Furthermore, LPS induced an increase in expression of mitophagy related proteins, PTEN-induced putative kinase (PINK1) and Parkin in the intestinal mitochondria, as well as an enhancement of the ratio of light chain 3-II (LC3-II) to LC3-I content in the jejunal mucosa. These results suggested that LPS-induced intestinal injury accompanied by disrupted antioxidant homeostasis, caused mt dysfunction and triggered mitophagy.
Collapse
Affiliation(s)
- Shuting Cao
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Qianhui Zhang
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - ChunChun Wang
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Huan Wu
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Lefei Jiao
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Qihua Hong
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Caihong Hu
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| |
Collapse
|
24
|
Qi J, Qian K, Tian L, Cheng Z, Wang Y. Gallium(iii)–2-benzoylpyridine-thiosemicarbazone complexes promote apoptosis through Ca2+ signaling and ROS-mediated mitochondrial pathways. NEW J CHEM 2018. [DOI: 10.1039/c8nj00697k] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ga(iii) complexes promoted apoptosis as a result of a combination of multiple apoptotic pathways.
Collapse
Affiliation(s)
- Jinxu Qi
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| | - Kun Qian
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| | - Liang Tian
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| | - Zhen Cheng
- Stanford Cancer Institute
- Member of Academic Council
- Stanford University
- USA
| | - Yihong Wang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| |
Collapse
|
25
|
Wang Y, Yu X, Song H, Feng D, Jiang Y, Wu S, Geng J. The STAT-ROS cycle extends IFN‑induced cancer cell apoptosis. Int J Oncol 2017; 52:305-313. [PMID: 29115415 DOI: 10.3892/ijo.2017.4196] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/30/2017] [Indexed: 11/06/2022] Open
Abstract
In mammals, the signal transducer and activator of transcription (STAT) protein processes mitochondria importation targets and mitochondria respiratory complexes, and triggers reactive oxygen species (ROS) generation, which conversely rapidly initiates the activation of STAT. Interferon (IFN) administration increases cancer cell apoptosis via STAT activation and ROS accumulation. However, the existence of a STAT-ROS cycle and how it affects IFN‑induced cancer cellular apoptosis are unclear. In the present study, we used MCF7 breast cancer cells and confirmed that a combination of IFN‑α/β/γ incubation induced STAT1/3 phosphorylation and mitochondria importation, which increased mitochondria respiratory complexes, the cellular oxygen consumption rate (OCR), and ROS production, followed by cellular apoptosis. We also found that STAT1/3 overexpression induced mitochondria respiratory complexes and ROS production. Additionally, ROS induced by H2O2 induced phosphorylation of STAT1/3 and promoted mitochondria importation. STAT1/3 deletion suppressed H2O2-induced acute cellular OCR, increasing the ROS level and indicating that STAT1/3 is necessary for ROS-induced mitochondria OCR and further ROS production, suggesting the existence of a STAT-ROS cycle. We next found that IFN induced mitochondria respiratory complexes followed by induction of OCR, ROS, and apoptosis, which were partially blocked by STAT1/3 deletion. Additionally, the suppression of ROS inhibited IFN‑induced STAT1/3 activation. Finally, we discovered that this cycle exists also in A431 and HeLa cancer cells. These results indicate that a STAT-ROS cycle extends IFN‑induced cellular apoptosis.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoyu Yu
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Hongtao Song
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Di Feng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Yang Jiang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Shuang Wu
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Jingshu Geng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
26
|
Deng SY, Zhang LM, Ai YH, Pan PH, Zhao SP, Su XL, Wu DD, Tan HY, Zhang LN, Tsung A. Role of interferon regulatory factor-1 in lipopolysaccharide-induced mitochondrial damage and oxidative stress responses in macrophages. Int J Mol Med 2017; 40:1261-1269. [PMID: 28849179 PMCID: PMC5593470 DOI: 10.3892/ijmm.2017.3110] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/04/2017] [Indexed: 01/20/2023] Open
Abstract
Sepsis causes many early deaths; both macrophage mitochondrial damage and oxidative stress responses are key factors in its pathogenesis. Although the exact mechanisms responsible for sepsis-induced mitochondrial damage are unknown, the nuclear transcription factor, interferon regulatory factor-1 (IRF-1) has been reported to cause mitochondrial damage in several diseases. Previously, we reported that in addition to promoting systemic inflammation, IRF-1 promoted the apoptosis of and inhibited autophagy in macrophages. In the present study, we hypothesized that lipopolysaccharide (LPS)-induced IRF-1 activation in macrophages may promote mitochondrial damage and oxidative stress. In vitro, LPS was found to promote IRF-1 activation, reactive oxygen species (ROS) production, adenosine triphosphate (ATP) depletion, superoxide dismutase (SOD) consumption, malondialdehyde (MDA) accumulation and mitochondrial depolarization in macrophages in a time- and dose-dependent manner. These effects were abrogated in cells in which IRF-1 was knocked down. Furthermore, IRF-1 overexpression increased LPS-induced oxidative stress responses and mitochondrial damage. In vivo, peritoneal macrophages obtained from IRF-1 knockout (KO) mice produced less ROS and had less mitochondrial depolarization and damage following the administration of LPS, when compared to their wild-type (WT) counterparts. In addition, IRF-1 KO mice exhibited a decreased release of mitochondrial DNA (mtDNA) following the administration of LPS. Thus, IRF-1 may be a critical factor in augmenting LPS-induced oxidative stress and mitochondrial damage in macrophages.
Collapse
Affiliation(s)
- Song-Yun Deng
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Le-Meng Zhang
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu-Hang Ai
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pin-Hua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shuang-Ping Zhao
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiao-Li Su
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Dong-Dong Wu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hong-Yi Tan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Li-Na Zhang
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
27
|
Lee SB, Kang JW, Kim SJ, Ahn J, Kim J, Lee SM. Afzelin ameliorates D-galactosamine and lipopolysaccharide-induced fulminant hepatic failure by modulating mitochondrial quality control and dynamics. Br J Pharmacol 2016; 174:195-209. [PMID: 27861739 DOI: 10.1111/bph.13669] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 11/01/2016] [Accepted: 11/08/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Fulminant hepatic failure (FHF) is a fatal clinical syndrome that results in excessive inflammation and hepatocyte death. Mitochondrial dysfunction is considered to be a possible mechanism of FHF. Afzelin, a flavonol glycoside found in Houttuynia cordata Thunberg, has anti-inflammatory and antioxidant properties. The present study elucidated the cytoprotective mechanisms of afzelin against D-galactosamine (GalN)/LPS induced FHF, particularly focusing on mitochondrial quality control and dynamics. EXPERIMENTAL APPROACH Mice were administered afzelin i.p. 1 h before receiving GalN (800 mg·kg-1 )/LPS (40 μg·kg-1 ), and they were then killed 5 h after GalN/LPS treatment. KEY RESULTS Afzelin improved the survival rate and reduced the serum levels of alanine aminotransferase and pro-inflammatory cytokines in GalN/LPS-treated mice. Afzelin attenuated the mitochondrial damage, as indicated by diminished mitochondrial swelling and mitochondrial glutamate dehydrogenase activity in GalN/LPS-treated mice. Afzelin enhanced mitochondrial biogenesis, as indicated by increased levels of PPAR-γ coactivator 1α, nuclear respiratory factor 1 and mitochondrial transcription factor A. Afzelin also decreased the level of mitophagy-related proteins, parkin and PTEN-induced putative kinase 1. Furthermore, while GalN/LPS significantly increased the level of fission-related protein, dynamin-related protein 1, and decreased the level of fusion-related protein, mitofusin 2; these effects were attenuated by afzelin. CONCLUSIONS AND IMPLICATIONS Our findings demonstrated that afzelin protects against GalN/LPS-induced liver injury by enhancing mitochondrial biogenesis, suppressing excessive mitophagy and balancing mitochondrial dynamics.
Collapse
Affiliation(s)
- Sang-Bin Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Jung-Woo Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - So-Jin Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Jongmin Ahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Jinwoong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
28
|
Xia X, Fu J, Song X, Shi Q, Su C, Song E, Song Y. Neohesperidin dihydrochalcone down-regulates MyD88-dependent and -independent signaling by inhibiting endotoxin-induced trafficking of TLR4 to lipid rafts. Free Radic Biol Med 2015; 89:522-32. [PMID: 26453923 DOI: 10.1016/j.freeradbiomed.2015.08.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 02/07/2023]
Abstract
Fulminant hepatic failure (FHF) is a lethal clinical syndrome characterized by the activation of macrophages and the increased production of inflammatory mediators. The purpose of this study was to investigate the effects of neohesperidin dihydrochalcone (NHDC), a widely-used low caloric artificial sweetener against FHF. An FHF experimental model was established in mice by intraperitoneal injection of D-galactosamine (d-GalN) (400mg/kg)/lipopolysaccharides (LPS) (10 μg/kg). Mice were orally administered NHDC for 6 continuous days and at 1h before d-GalN/LPS administration. RAW264.7 macrophages were used as an in vitro model. Cells were pre-treated with NHDC for 1h before stimulation with LPS (10 μg/ml) for 6h. d-GalN/LPS markedly increased the serum transaminase activities and levels of oxidative and inflammatory markers, which were significantly attenuated by NHDC. Mechanistic analysis indicated that NHDC inhibited LPS-induced myeloid differentiation factor 88 (MyD88) and TIR-containing adapter molecule (TRIF)-dependent signaling. Transient transfection of TLR4 or MyD88 siRNA inhibited the downstream inflammatory signaling. This effect could also be achieved by the pretreatment with NHDC. The fluorescence microscopy and flow cytometry results suggested that NHDC potently inhibited the binding of LPS to TLR4 in RAW264.7 macrophages. In addition, the inhibitory effect of NHDC on LPS-induced translocation of TLR4 into lipid raft domains played an important role in the amelioration of production of downstream pro-inflammatory molecules. Furthermore, the activation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) by NHDC inhibited TLR4 signaling. In conclusion, our results suggest that NHDC attenuates d-GalN/LPS-induced FHF by inhibiting the TLR4-mediated inflammatory pathway, demonstrating a new application of NHDC as a hepatoprotective agent.
Collapse
Affiliation(s)
- Xiaomin Xia
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Juanli Fu
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Xiufang Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Qiong Shi
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Chuanyang Su
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Erqun Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Yang Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715.
| |
Collapse
|
29
|
Dose response of endotoxin on hepatocyte and muscle mitochondrial respiration in vitro. BIOMED RESEARCH INTERNATIONAL 2015; 2015:353074. [PMID: 25649304 PMCID: PMC4306363 DOI: 10.1155/2015/353074] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/22/2014] [Accepted: 09/08/2014] [Indexed: 01/02/2023]
Abstract
Introduction. Results on mitochondrial dysfunction in sepsis are controversial. We aimed to assess effects of LPS at wide dose and time ranges on hepatocytes and isolated skeletal muscle mitochondria. Methods. Human hepatocellular carcinoma cells (HepG2) were exposed to placebo or LPS (0.1, 1, and 10 μg/mL) for 4, 8, 16, and 24 hours and primary human hepatocytes to 1 μg/mL LPS or placebo (4, 8, and 16 hours). Mitochondria from porcine skeletal muscle samples were exposed to increasing doses of LPS (0.1–100 μg/mg) for 2 and 4 hours. Respiration rates of intact and permeabilized cells and isolated mitochondria were measured by high-resolution respirometry. Results. In HepG2 cells, LPS reduced mitochondrial membrane potential and cellular ATP content but did not modify basal respiration. Stimulated complex II respiration was reduced time-dependently using 1 μg/mL LPS. In primary human hepatocytes, stimulated mitochondrial complex II respiration was reduced time-dependently using 1 μg/mL LPS. In isolated porcine skeletal muscle mitochondria, stimulated respiration decreased at high doses (50 and 100 μg/mL LPS). Conclusion. LPS reduced cellular ATP content of HepG2 cells, most likely as a result of the induced decrease in membrane potential. LPS decreased cellular and isolated mitochondrial respiration in a time-dependent, dose-dependent and complex-dependent manner.
Collapse
|
30
|
Seo J, Lee JY, Sung MS, Byun CJ, Cho DH, Lee HJ, Park JH, Cho HS, Cho SJ, Jo I. Arsenite Acutely Decreases Nitric Oxide Production via the ROS-Protein Phosphatase 1-Endothelial Nitric Oxide Synthase-Thr(497) Signaling Cascade. Biomol Ther (Seoul) 2014; 22:510-8. [PMID: 25489418 PMCID: PMC4256030 DOI: 10.4062/biomolther.2014.106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 11/17/2022] Open
Abstract
Chronic (>24 h) exposure of arsenite, an environmental toxicant, has shown the decreased nitric oxide (NO) production in endothelial cells (EC) by decreasing endothelial NO synthase (eNOS) expression and/or its phosphorylation at serine 1179 (eNOS-Ser1179 in bovine sequence), which is associated with increased risk of vascular diseases. Here, we investigated the acute (<24 h) effect of arsenite on NO production using bovine aortic EC (BAEC). Arsenite acutely increased the phosphorylation of eNOS-Thr497, but not of eNOS-Ser116 or eNOS-Ser1179, which was accompanied by decreased NO production. The level of eNOS expression was unaltered under this condition. Treatment with arsenite also induced reactive oxygen species (ROS) production, and pretreatment with a ROS scavenger N-acetyl-L-cysteine (NAC) completely reversed the observed effect of arsenite on eNOS-Thr497 phosphorylation. Although protein kinase C (PKC) and protein phosphatase 1 (PP1) were reported to be involved in eNOS-Thr497 phosphorylation, treatment with PKC inhibitor, Ro318425, and overexpression of various PKC isoforms did not affect the arsenite-stimulated eNOS-Thr497 phosphorylation. In contrast, treatment with PP1 inhibitor, calyculin A, mimicked the observed effect of arsenite on eNOS-Thr497 phosphorylation. Lastly, we found decreased cellular PP1 activity in arsenite-treated cells, which was reversed by NAC. Overall, our study demonstrates firstly that arsenite acutely decreases NO production at least in part by increasing eNOS-Thr497 phosphorylation via ROS-PP1 signaling pathway, which provide the molecular mechanism underlying arsenite-induced increase in vascular disease.
Collapse
Affiliation(s)
- Jungwon Seo
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710 ; Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan 570-749
| | - Jee Young Lee
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | - Min-Sun Sung
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | | | - Du-Hyong Cho
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 301-768
| | - Hyeon-Ju Lee
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | - Jung-Hyun Park
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | - Ho-Seong Cho
- Biosafety Research Institute and College of Veterinary Medicine, Chonbuk National University, Jeonju 561-756
| | - Sung-Jin Cho
- Department of Biology, College of Natural Sciences, Chungbuk National University, Cheongju 362-763, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| |
Collapse
|
31
|
The regulation role of interferon regulatory factor-1 gene and clinical relevance. Hum Immunol 2014; 75:1110-4. [DOI: 10.1016/j.humimm.2014.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 11/20/2022]
|
32
|
Zhang J, Xu L, Zhang L, Ying Z, Su W, Wang T. Curcumin attenuates D-galactosamine/lipopolysaccharide-induced liver injury and mitochondrial dysfunction in mice. J Nutr 2014; 144:1211-8. [PMID: 24899159 DOI: 10.3945/jn.114.193573] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Curcumin, a naturally occurring antioxidant, has various beneficial effects in the treatment of human diseases. However, little information regarding the protection it provides against acute liver injury is available. The present study investigated the protective effects of curcumin against D-galactosamine (D-GalN)/lipopolysaccharide (LPS)-induced acute liver injury in mice. A total of 40 male Kunming mice were randomly assigned to 5 groups: 1) mice administered saline vehicle injection (control), 2) mice administered 200 mg/kg body weight (BW) curcumin by i.p. injection (CUR), 3) mice administered D-GalN/LPS (700 mg and 5 μg/kg BW) via i.p. injection (GL), 4) mice administered 200 mg/kg BW curcumin i.p. 1 h before D-GalN/LPS injection (CUR-GL), and 5) mice administered 200 mg/kg BW curcumin i.p. 1 h after D-GalN/LPS injection (GL-CUR). Twenty h after D-GalN/LPS injection, serum alanine aminotransferase activities were 18.5% and 13.5% lower (P < 0.05) and aspartate aminotransferase (AST) activities were 26.6% and 9.6% lower (P < 0.05) in the CUR-GL and GL-CUR groups, respectively, than in the GL group. The CUR-GL and GL-CUR groups had 64.4% and 15.0% higher (P < 0.05) mitochondrial membrane potentials, respectively, and the CUR-GL group had a 44.7% lower reactive oxygen species concentration than the GL group (P < 0.05). Mitochondrial manganese superoxide dismutase activities were 111% and 77.9% higher (P < 0.05) and the percentages of necrotic cells were 47.0% and 32.4% lower (P < 0.05) in the CUR-GL and GL-CUR groups, respectively, than in the GL group. Liver mRNA levels of sirtuin 1 (Sirt1) were 56.4% lower (P < 0.05) in the CUR-GL group than in the GL group. Moreover, compared with the GL-CUR group, the CUR-GL group had an 18.7% lower serum AST activity, a 31.7% lower mitochondrial malondialdehyde concentration, a 36.0% lower hepatic reactive oxygen species concentration, and a 43.0% higher mitochondrial membrane potential. These results suggested that curcumin protects against D-GalN/LPS-induced liver damage by the enhancing antioxidant defense system, attenuating mitochondrial dysfunction and inhibiting apoptosis. This was especially true for curcumin pretreatment, which highlighted its promise as a preventive treatment for acute liver injury in clinical settings.
Collapse
Affiliation(s)
- Jingfei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Li Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Zhixiong Ying
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Weipeng Su
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| |
Collapse
|
33
|
XUE LI, LI MING, CHEN TENG, SUN HAIFENG, ZHU JIE, LI XIA, WU FENG, WANG BIAO, LI JUPING, CHEN YANJIONG. PE‑induced apoptosis in SMMC‑7721 cells: involvement of Erk and Stat signalling pathways. Int J Mol Med 2014; 34:119-29. [PMID: 24821075 PMCID: PMC4072400 DOI: 10.3892/ijmm.2014.1777] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/30/2014] [Indexed: 12/16/2022] Open
Abstract
Emerging evidence indicates that the redistribution of phosphatidylethanolamine (PE) across the bilayer of the plasma membrane is an important molecular marker for apoptosis. However, the effect of PE on apoptosis and the underlying mechanism of PE remain unclear. In the current study, MTT and flow cytometric assays were used to examine the effects of PE on apoptosis in SMMC‑7721 cells. The level of mitochondrial membrane potential (ΔΨm) and the expression of Bax, Bcl‑2, caspase‑3, phospho‑Erk and phospho‑Stat1/2 in SMMC‑7721 cells that were exposed to PE were also investigated. The results showed that PE inhibited proliferation, caused G0/G1 phase cell cycle arrest and induced apoptosis in SMMC‑7721 cells in a dose‑dependent manner. Rhodamine 123 staining showed that the treatment of SMMC‑7721 cells with different concentrations of PE for 24 h significantly decreased the level of ΔΨm and exerted dose‑dependent effects. Using immunofluorescence and western blotting, we found that the expression of Bax was upregulated, whereas that of Bcl‑2 was downregulated in PE‑induced apoptotic cells. In addition, these events were accompanied by an increase in caspase‑3 expression in a dose‑dependent manner following PE treatment. PE‑induced apoptosis was accompanied by a decrease in Erk phospho-rylation and by the activation of Stat1/2 phosphorylation in SMMC‑7721 cells. In conclusion, the results suggested that PE‑induced apoptosis is involved in upregulating the Bax/Bcl‑2 protein ratio and decreasing the ΔΨm. Moreover, the results showed that the Erk and Stat1/2 signalling pathways may be involved in the process of PE‑induced apoptosis.
Collapse
Affiliation(s)
- LI XUE
- Forensic Medicine College of Xi’an Jiaotong University, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of the Ministry of Education for Environment and Genes Related to Diseases, Xi’an, Shaanxi 710061, P.R. China
- Department of Immunology and Pathogenic Biology, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi 710061, P.R. China
| | - MING LI
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, P.R. China
| | - TENG CHEN
- Forensic Medicine College of Xi’an Jiaotong University, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of the Ministry of Education for Environment and Genes Related to Diseases, Xi’an, Shaanxi 710061, P.R. China
| | - HAIFENG SUN
- Tumour Hospital of Shaanxi Province, Xi’an, Shaanxi 710061, P.R. China
| | - JIE ZHU
- Forensic Medicine College of Xi’an Jiaotong University, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of the Ministry of Education for Environment and Genes Related to Diseases, Xi’an, Shaanxi 710061, P.R. China
| | - XIA LI
- VIP Internal Medicine Department, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - FENG WU
- Graduate Teaching and Experimental Centre, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi 710061, P.R. China
| | - BIAO WANG
- Department of Immunology and Pathogenic Biology, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi 710061, P.R. China
| | - JUPING LI
- School of Public Security, Northwest University of Politics and Law, Xi’an, Shaanxi 710063, P.R. China
| | - YANJIONG CHEN
- Department of Immunology and Pathogenic Biology, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi 710061, P.R. China
| |
Collapse
|
34
|
Abstract
All seven STAT proteins are expressed in the heart, and in this review we will focus on their contribution to cardiac physiology and to ischemic heart disease and its consequences. A substantial literature has focused on the roles of STAT1 and STAT3 in ischemic heart disease, where, at least in the acute phase, they appear to have a yin-yang relationship. STAT1 contributes to the loss of irreplaceable cardiac myocytes both by increasing apoptosis and by reducing cardioprotective autophagy. In contrast, STAT3 is cardioprotective, since STAT3-deficient mice have larger infarcts following ischemic injury, and a number of cardioprotective agents have been shown to act, at least partly, through STAT3 activation. STAT3 is also absolutely required for preconditioning—a process where periods of brief ischemia protect against a subsequent or previous prolonged ischemic episode. Prolonged activation of STAT3, however, is strongly implicated in the post-infarction remodeling of the heart which leads to heart failure, where, possibly together with STAT5, it augments activation of the renin-angiotensin system.
Collapse
Affiliation(s)
- Richard A Knight
- Medical Molecular Biology Unit; University College London; London, UK
| | | | | |
Collapse
|
35
|
Meier JA, Larner AC. Toward a new STATe: the role of STATs in mitochondrial function. Semin Immunol 2014; 26:20-8. [PMID: 24434063 PMCID: PMC4321820 DOI: 10.1016/j.smim.2013.12.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 12/23/2013] [Indexed: 12/11/2022]
Abstract
Signal Transducers and Activators of Transcription (STATs) have been studied extensively and have been associated with virtually every biochemical pathway. Until recently, however, they were thought to exert these effects solely as a nuclear transcription factor. The finding that STAT3 localizes to the mitochondria and modulates respiration has opened up a new avenue through which STATs may regulate the cell. Recently, other members of the STAT family (STAT1, STAT2, STAT5, and STAT6) have also been shown to be present in the mitochondria. Coordinate regulation at the nucleus and mitochondria by these proteins places them in a unique position to drive cellular processes to achieve a specific response. This review summarizes recent findings that have led to our current understanding of how STATs influence mitochondrial function in health and disease.
Collapse
Affiliation(s)
- Jeremy A. Meier
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA,Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Andrew C. Larner
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA,Corresponding author at: Department of Biochemistry and Molecular Biology, and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA. Tel.: +1 804 828 2903; fax: +1 804 827 1657. (A.C. Larner)
| |
Collapse
|
36
|
Abstract
TNF-induced necroptosis is caused by the activation of RIPK1 and the subsequent production of reactive oxygen species in the mitochondria, although the intermittent molecules of the signaling pathway responsible for this ROS-mediated type of programmed necrosis have not yet been identified. A recent article by Shulga and Pastorino in the Journal of Cell Science identifies RIPK1 as the mediator of STAT3 Ser727 phosphorylation, which leads to the translocation of the latter into the mitochondria via its interaction with GRIM-19, a member of the mitochondrial complex I. Here we discuss how the findings of the Shulga and Pastorino study shed light onto the involvement of STAT3 in necroptosis.
Collapse
|
37
|
Kurian GA, Berenshtein E, Kakhlon O, Chevion M. Energy status determines the distinct biochemical and physiological behavior of interfibrillar and sub-sarcolemmal mitochondria. Biochem Biophys Res Commun 2012; 428:376-82. [DOI: 10.1016/j.bbrc.2012.10.062] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 10/16/2012] [Indexed: 12/30/2022]
|
38
|
Lin ST, Chou HC, Chang SJ, Chen YW, Lyu PC, Wang WC, Chang MDT, Chan HL. Proteomic analysis of proteins responsible for the development of doxorubicin resistance in human uterine cancer cells. J Proteomics 2012; 75:5822-47. [DOI: 10.1016/j.jprot.2012.07.047] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 07/16/2012] [Accepted: 07/20/2012] [Indexed: 10/28/2022]
|
39
|
Li P, Zhao Y, Wu X, Xia M, Fang M, Iwasaki Y, Sha J, Chen Q, Xu Y, Shen A. Interferon gamma (IFN-γ) disrupts energy expenditure and metabolic homeostasis by suppressing SIRT1 transcription. Nucleic Acids Res 2011; 40:1609-20. [PMID: 22064865 PMCID: PMC3287208 DOI: 10.1093/nar/gkr984] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Chronic inflammation impairs metabolic homeostasis and is intimately correlated with the pathogenesis of type 2 diabetes. The pro-inflammatory cytokine IFN-γ is an integral part of the metabolic inflammation circuit and contributes significantly to metabolic dysfunction. The underlying mechanism, however, remains largely unknown. In the present study, we report that IFN-γ disrupts the expression of genes key to cellular metabolism and energy expenditure by repressing the expression and activity of SIRT1 at the transcription level. Further analysis reveals that IFN-γ requires class II transactivator (CIITA) to repress SIRT1 transcription. CIITA, once induced by IFN-γ, is recruited to the SIRT1 promoter by hypermethylated in cancer 1 (HIC1) and promotes down-regulation of SIRT1 transcription via active deacetylation of core histones surrounding the SIRT1 proximal promoter. Silencing CIITA or HIC1 restores SIRT1 activity and expression of metabolic genes in skeletal muscle cells challenged with IFN-γ. Therefore, our data delineate an IFN-γ/HIC1/CIITA axis that contributes to metabolic dysfunction by suppressing SIRT1 transcription in skeletal muscle cells and as such shed new light on the development of novel therapeutic strategies against type 2 diabetes.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Reproductive Medicine and Department of Pathophysiology, Key Laboratory of Cardiovascular Disease,The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Singh P, Goode T, Dean A, Awad SS, Darlington GJ. Elevated interferon gamma signaling contributes to impaired regeneration in the aged liver. J Gerontol A Biol Sci Med Sci 2011; 66:944-56. [PMID: 21719609 DOI: 10.1093/gerona/glr094] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our previous study on immune-related changes in the aged liver described immune cell infiltration and elevation of inflammation with age. Levels of interferon (IFN)-γ, a known cell cycle inhibitor, were elevated in the aging liver. Here, we determine the role played by IFN-γ in the delayed regenerative response observed in the aged livers. We observed elevated IFN signaling in both aged hepatocytes and regenerating livers post-partial hepatectomy. In vivo deletion of the major IFN-γ producers-the macrophages and the natural killer cells, leads to a reduction in the IFN-γ levels accompanied with the restoration of the DNA synthesis kinetics in the aged livers. Eighteen-month-old IFN-γ-/- mice livers, upon resection, exhibited an earlier entry into the cell cycle compared with age-matched controls. Thus, our study strongly suggests that an age-related elevation in inflammatory conditions in the liver often dubbed as "inflammaging" has a detrimental effect on the regenerative response.
Collapse
Affiliation(s)
- Pallavi Singh
- Department of Dermatology, Columbia University Medical Center, 1150 St. Nicholas Avenue, New York, NY 10032, USA.
| | | | | | | | | |
Collapse
|
41
|
Lin HW, Thompson JW, Morris KC, Perez-Pinzon MA. Signal transducers and activators of transcription: STATs-mediated mitochondrial neuroprotection. Antioxid Redox Signal 2011; 14:1853-61. [PMID: 20712401 PMCID: PMC3078497 DOI: 10.1089/ars.2010.3467] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 08/02/2010] [Accepted: 08/13/2010] [Indexed: 01/07/2023]
Abstract
Cerebral ischemia is defined as little or no blood flow in cerebral circulation, characterized by low tissue oxygen and glucose levels, which promotes neuronal mitochondria dysfunction leading to cell death. A strategy to counteract cerebral ischemia-induced neuronal cell death is ischemic preconditioning (IPC). IPC results in neuroprotection, which is conferred by a mild ischemic challenge prior to a normally lethal ischemic insult. Although many IPC-induced mechanisms have been described, many cellular and subcellular mechanisms remain undefined. Some reports have suggested key signal transduction pathways of IPC, such as activation of protein kinase C epsilon, mitogen-activated protein kinase, and hypoxia-inducible factors, that are likely involved in IPC-induced mitochondria mediated-neuroprotection. Moreover, recent findings suggest that signal transducers and activators of transcription (STATs), a family of transcription factors involved in many cellular activities, may be intimately involved in IPC-induced ischemic tolerance. In this review, we explore current signal transduction pathways involved in IPC-induced mitochondria mediated-neuroprotection, STAT activation in the mitochondria as it relates to IPC, and functional significance of STATs in cerebral ischemia.
Collapse
Affiliation(s)
- Hung Wen Lin
- Cerebral Vascular Disease Research Center, Department of Neurology, University of Miami, Miller School of Medicine, Miami, Florida 33101, USA
| | | | | | | |
Collapse
|
42
|
Depinay N, Franetich JF, Grüner AC, Mauduit M, Chavatte JM, Luty AJF, van Gemert GJ, Sauerwein RW, Siksik JM, Hannoun L, Mazier D, Snounou G, Rénia L. Inhibitory effect of TNF-α on malaria pre-erythrocytic stage development: influence of host hepatocyte/parasite combinations. PLoS One 2011; 6:e17464. [PMID: 21394207 PMCID: PMC3048870 DOI: 10.1371/journal.pone.0017464] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 01/25/2011] [Indexed: 11/24/2022] Open
Abstract
Background The liver stages of malaria parasites are inhibited by cytokines such as interferon-γ or Interleukin (IL)-6. Binding of these cytokines to their receptors at the surface of the infected hepatocytes leads to the production of nitric oxide (NO) and radical oxygen intermediates (ROI), which kill hepatic parasites. However, conflicting results were obtained with TNF-α possibly because of differences in the models used. We have reassessed the role of TNF-α in the different cellular systems used to study the Plasmodium pre-erythrocytic stages. Methods and Findings Human or mouse TNF-α were tested against human and rodent malaria parasites grown in vitro in human or rodent primary hepatocytes, or in hepatoma cell lines. Our data demonstrated that TNF-α treatment prevents the development of malaria pre-erythrocytic stages. This inhibitory effect however varies with the infecting parasite species and with the nature and origin of the cytokine and hepatocytes. Inhibition was only observed for all parasite species tested when hepatocytes were pre-incubated 24 or 48 hrs before infection and activity was directed only against early hepatic parasite. We further showed that TNF-α inhibition was mediated by a soluble factor present in the supernatant of TNF-α stimulated hepatocytes but it was not related to NO or ROI. Treatment TNF-α prevents the development of human and rodent malaria pre-erythrocytic stages through the activity of a mediator that remains to be identified. Conclusions Treatment TNF-α prevents the development of human and rodent malaria pre-erythrocytic stages through the activity of a mediator that remains to be identified. However, the nature of the cytokine-host cell-parasite combination must be carefully considered for extrapolation to the human infection.
Collapse
Affiliation(s)
- Nadya Depinay
- Institut Cochin, Département d'Immunologie, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
| | - Jean Francois Franetich
- INSERM UMRS 945, Paris, France
- Université Pierre & Marie Curie, Faculté de Médecine Pitié-Salpêtrière, Paris, France
| | - Anne Charlotte Grüner
- Institut Cochin, Département d'Immunologie, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Marjorie Mauduit
- Institut Cochin, Département d'Immunologie, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Jean-Marc Chavatte
- Equipe Parasitologie Comparée et Modèles Expérimentaux USM0307, CNRS IFR101, Muséum National d'Histoire Naturelle, Paris, France
| | - Adrian J. F. Luty
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Robert W. Sauerwein
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jean-Michel Siksik
- Assistance Publique-Hôpitaux de Paris, Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Laurent Hannoun
- Assistance Publique-Hôpitaux de Paris, Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Dominique Mazier
- INSERM UMRS 945, Paris, France
- Université Pierre & Marie Curie, Faculté de Médecine Pitié-Salpêtrière, Paris, France
- AP HP, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Georges Snounou
- INSERM UMRS 945, Paris, France
- Université Pierre & Marie Curie, Faculté de Médecine Pitié-Salpêtrière, Paris, France
- Equipe Parasitologie Comparée et Modèles Expérimentaux USM0307, CNRS IFR101, Muséum National d'Histoire Naturelle, Paris, France
- AP HP, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Laurent Rénia
- Institut Cochin, Département d'Immunologie, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
- * E-mail:
| |
Collapse
|
43
|
Yang IV, Alper S, Lackford B, Rutledge H, Warg LA, Burch LH, Schwartz DA. Novel regulators of the systemic response to lipopolysaccharide. Am J Respir Cell Mol Biol 2010; 45:393-402. [PMID: 21131441 DOI: 10.1165/rcmb.2010-0342oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Our understanding of the role that host genetic factors play in the initiation and severity of infections caused by gram-negative bacteria is incomplete. To identify novel regulators of the host response to lipopolysaccharide (LPS), 11 inbred murine strains were challenged with LPS systemically. In addition to two strains lacking functional TLR4 (C3H/HeJ and C57BL/6J(TLR4-/-)), three murine strains with functional TLR4 (C57BL/6J, 129/SvImJ, and NZW/LacJ) were found to be relatively resistant to systemic LPS challenge; the other six strains were classified as sensitive. RNA from lung, liver, and spleen tissue was profiled on oligonucleotide microarrays to determine if unique transcripts differentiate susceptible and resistant strains. Gene expression analysis identified the Hedgehog signaling pathway and a number of transcription factors (TFs) involved in the response to LPS. RNA interference-mediated inhibition of six TFs (C/EBP, Cdx-2, E2F1, Hoxa4, Nhlh1, and Tead2) was found to diminish IL-6 and TNF-α production by murine macrophages. Mouse lines with targeted mutations were used to verify the involvement of two novel genes in innate immunity. Compared with wild-type control mice, mice deficient in the E2F1 transcription factor were found to have a reduced inflammatory response to systemic LPS, and mice heterozygote for Ptch, a gene involved in Hedgehog signaling, were found to be more responsive to systemic LPS. Our analysis of gene expression data identified novel pathways and transcription factors that regulate the host response to systemic LPS. Our results provide potential sepsis biomarkers and therapeutic targets that should be further investigated in human populations.
Collapse
Affiliation(s)
- Ivana V Yang
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Kim JY, Song EH, Lee HJ, Oh YK, Park YS, Park JW, Kim BJ, Kim DJ, Lee I, Song J, Kim WH. Chronic ethanol consumption-induced pancreatic {beta}-cell dysfunction and apoptosis through glucokinase nitration and its down-regulation. J Biol Chem 2010; 285:37251-62. [PMID: 20855893 DOI: 10.1074/jbc.m110.142315] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic ethanol consumption is known as an independent risk factor for type 2 diabetes, which is characterized by impaired glucose homeostasis and insulin resistance; however, there is a great deal of controversy concerning the relationships between alcohol consumption and the development of type 2 diabetes. We investigated the effects of chronic ethanol consumption on pancreatic β-cell dysfunction and whether generated peroxynitrite participates in the impaired glucose homeostasis. Here we show that chronic ethanol feeding decreases the ability of pancreatic β-cells to mediate insulin secretion and ATP production in coordination with the decrease of glucokinase, Glut2, and insulin expression. Specific blockade of ATF3 using siRNA or C-terminally deleted ATF3(ΔC) attenuated ethanol-induced pancreatic β-cell apoptosis or dysfunction and restored the down-regulation of glucokinase (GCK), insulin, and pancreatic duodenal homeobox-1 induced by ethanol. GCK inactivation and down-regulation were predominantly mediated by ethanol metabolism-generated peroxynitrite, which were suppressed by the peroxynitrite scavengers N(γ)-monomethyl-L-arginine, uric acid, and deferoxamine but not by the S-nitrosylation inhibitor DTT, indicating that tyrosine nitration is the predominant modification associated with GCK down-regulation and inactivation rather than S-nitrosylation of cysteine. Tyrosine nitration of GCK prevented its association with pBad, and GCK translocation into the mitochondria results in subsequent proteasomal degradation of GCK following ubiquitination. This study identified a novel and efficient pathway by which chronic ethanol consumption may induce GCK down-regulation and inactivation by inducing tyrosine nitration of GCK, resulting in pancreatic β-cell apoptosis and dysfunction. Peroxynitrite-induced ATF3 may also serve as a potent upstream regulator of GCK down-regulation and β-cell apoptosis.
Collapse
Affiliation(s)
- Ji Yeon Kim
- Divisions of Metabolic Diseases, National Institutes of Health, Eunpyeong-gu, Seoul 122-701, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ueki S, Dhupar R, Cardinal J, Tsung A, Yoshida J, Ozaki KS, Klune JR, Murase N, Geller DA. Critical role of interferon regulatory factor-1 in murine liver transplant ischemia reperfusion injury. Hepatology 2010; 51:1692-701. [PMID: 20131404 PMCID: PMC3001118 DOI: 10.1002/hep.23501] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Interferon regulatory factor-1 (IRF-1) is a transcription factor that regulates gene expression during immunity. We hypothesized that IRF-1 plays a pivotal role in liver transplant (LTx) ischemia/reperfusion (I/R) injury. Mouse orthotopic LTx was conducted after 24 hours cold storage in University of Wisconsin (UW) solution in wildtype (WT) C57BL/6 and IRF-1 knockout (KO) mice. IRF-1 deficiency in liver grafts, but not in recipients, resulted in significant reduction of hepatocyte apoptosis and liver injury, as well as improved survival. IRF-1 mRNA up-regulation was typically seen in graft hepatocytes in WT-->WT LTx. Deficiency of IRF-1 signaling in graft resulted in significantly reduced messenger RNA (mRNA) levels for death ligands and death receptors in hepatocytes, as well as decreased caspase-8 activities, indicating that IRF-1 mediates death ligand-induced hepatocyte death. Further, a smaller but significant IRF-1 mRNA up-regulation was seen in WT graft nonparenchymal cells (NPC) and associated with interferon gamma (IFN-gamma) mRNA up-regulation exclusively in NPC. IFN-gamma mRNA was significantly reduced in IRF-1 KO graft. Thus, IRF-1 in graft hepatocytes and NPC has distinct effects in hepatic I/R injury. However, LTx with chimeric liver grafts showed that grafts lacking hepatocellular IRF-1 had better protection compared with those lacking IRF-1 in NPC. The study identifies a critical role for IRF-1 in liver transplant I/R injury.
Collapse
Affiliation(s)
- Shinya Ueki
- Thomas E. Starzl Transplant Institute, University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - Rajeev Dhupar
- University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - Jon Cardinal
- University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - Allan Tsung
- University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - Junichi Yoshida
- Thomas E. Starzl Transplant Institute, University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - Kikumi S. Ozaki
- Thomas E. Starzl Transplant Institute, University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - John Robert Klune
- University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - Noriko Murase
- Thomas E. Starzl Transplant Institute, University of Pittsburgh, Department of Surgery, Pittsburgh PA, 15213
| | - David A. Geller
- University of Pittsburgh Medical Center, Liver Cancer Center.,Corresponding Author: David A. Geller, M.D., UPMC Liver Cancer Center, 3471 Fifth Avenue, Kaufmann Medical Building, Suite 300, Pittsburgh, PA 15213. Phone: 412-692-2001; Fax: 412-692-2002;
| |
Collapse
|
46
|
Kim JY, Lee SH, Song EH, Park YM, Lim JY, Kim DJ, Choi KH, Park SI, Gao B, Kim WH. A critical role of STAT1 in streptozotocin-induced diabetic liver injury in mice: controlled by ATF3. Cell Signal 2009; 21:1758-67. [PMID: 19647793 PMCID: PMC4779502 DOI: 10.1016/j.cellsig.2009.07.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 07/18/2009] [Accepted: 07/18/2009] [Indexed: 02/07/2023]
Abstract
It is well-established that the administration of streptozotocin accelerates diabetic liver injury as well as type-I diabetes, however the underlying mechanisms are poorly understood. Here we investigated the molecular mechanisms of diabetic liver injury in a model of streptozotocin (STZ)-induced type-I diabetes. STZ administration induced type-1 diabetes and chronic liver injury was associated with increased STAT1, which is implicated in diabetic liver injury by virtue of its ability to promote hepatocyte apoptosis, in the liver and pancreas, which were all strongly inhibited in STAT1(-)(/-) mice. Similarly, STZ-induced ATF3, a stress-inducible gene, was completely abolished in the liver of IFN-gamma(-/-) mice, but not in STAT1(-/-) mice. Inhibition of STAT1 by siRNA or dominant-negative DNA did not affect ATF3 protein expression but blocked IFN-gamma-induced ATF3 translocation from the cytosol into the nucleus. In contrast, inhibition of ATF3 by using siRNA diminished STAT1 protein expression and IFN-gamma/STZ-induced hepatocyte apoptosis. Furthermore, GST pull-down and co-IP assay showed that STAT1 bound to C-terminal domain of ATF3. Such direct interaction increased the stability of STAT1 by inhibiting its ubiquitination as well as proteasome activity. Our results suggest that STAT1 is a common signaling pathway contributing to STZ-induced diabetes and diabetic liver injury. ATF3 functions as a potent regulator of STAT1 stability, accelerating STZ-induced diabetes and diabetic liver injury.
Collapse
Affiliation(s)
- Ji Yeon Kim
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institutes of Health, #194 Tongillo, Eunpyeong-gu, Seoul 122-701, Korea
| | - Sung Hee Lee
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institutes of Health, #194 Tongillo, Eunpyeong-gu, Seoul 122-701, Korea
| | - Eun Hyeon Song
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institutes of Health, #194 Tongillo, Eunpyeong-gu, Seoul 122-701, Korea
| | - Young Mi Park
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institutes of Health, #194 Tongillo, Eunpyeong-gu, Seoul 122-701, Korea
| | - Joong-Yeon Lim
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institutes of Health, #194 Tongillo, Eunpyeong-gu, Seoul 122-701, Korea
| | - Dae Jin Kim
- Department of Psychiatry, College of Medicine, Catholic University, Seoul, Korea
| | - Kyung-Hee Choi
- Laboratory of Molecular Biology, Department of Biology, College of Natural Sciences, Chung-Ang University, Seoul, Korea
| | - Sang Ick Park
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institutes of Health, #194 Tongillo, Eunpyeong-gu, Seoul 122-701, Korea
| | - Bin Gao
- Section on Liver Biology, Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, United States
| | - Won-Ho Kim
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institutes of Health, #194 Tongillo, Eunpyeong-gu, Seoul 122-701, Korea
| |
Collapse
|
47
|
Timmins JM, Ozcan L, Seimon TA, Li G, Malagelada C, Backs J, Backs T, Bassel-Duby R, Olson EN, Anderson ME, Tabas I. Calcium/calmodulin-dependent protein kinase II links ER stress with Fas and mitochondrial apoptosis pathways. J Clin Invest 2009; 119:2925-41. [PMID: 19741297 DOI: 10.1172/jci38857] [Citation(s) in RCA: 358] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 07/01/2009] [Indexed: 12/28/2022] Open
Abstract
ER stress-induced apoptosis is implicated in various pathological conditions, but the mechanisms linking ER stress-mediated signaling to downstream apoptotic pathways remain unclear. Using human and mouse cell culture and in vivo mouse models of ER stress-induced apoptosis, we have shown that cytosolic calcium resulting from ER stress induces expression of the Fas death receptor through a pathway involving calcium/calmodulin-dependent protein kinase IIgamma (CaMKIIgamma) and JNK. Remarkably, CaMKIIgamma was also responsible for processes involved in mitochondrial-dependent apoptosis, including release of mitochondrial cytochrome c and loss of mitochondrial membrane potential. CaMKII-dependent apoptosis was also observed in a number of cultured human and mouse cells relevant to ER stress-induced pathology, including cultured macrophages, endothelial cells, and neuronal cells subjected to proapoptotic ER stress. Moreover, WT mice subjected to systemic ER stress showed evidence of macrophage mitochondrial dysfunction and apoptosis, renal epithelial cell apoptosis, and renal dysfunction, and these effects were markedly reduced in CaMKIIgamma-deficient mice. These data support an integrated model in which CaMKII serves as a unifying link between ER stress and the Fas and mitochondrial apoptotic pathways. Our study also revealed what we believe to be a novel proapoptotic function for CaMKII, namely, promotion of mitochondrial calcium uptake. These findings raise the possibility that CaMKII inhibitors could be useful in preventing apoptosis in pathological settings involving ER stress-induced apoptosis.
Collapse
Affiliation(s)
- Jenelle M Timmins
- Department of Medicine, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Schmitt NC, Rubel EW, Nathanson NM. Cisplatin-induced hair cell death requires STAT1 and is attenuated by epigallocatechin gallate. J Neurosci 2009; 29:3843-51. [PMID: 19321781 PMCID: PMC2707781 DOI: 10.1523/jneurosci.5842-08.2009] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 01/07/2009] [Indexed: 12/20/2022] Open
Abstract
Cisplatin is a chemotherapy drug that frequently causes auditory impairment due to the death of mechanosensory hair cells. Cisplatin ototoxicity may result from oxidative stress, DNA damage, and inflammatory cytokines. The transcription factor STAT1, an important mediator of cell death, can regulate all of these processes in other cell types. We used cultured utricles from mature Swiss Webster mice to investigate the role of STAT1 in cisplatin-induced hair cell death. We show that STAT1 phosphorylation is an early event in both hair cells and support cells after exposure of utricles to cisplatin. STAT1 phosphorylation peaked after 4 h of cisplatin exposure and returned to control levels by 8 h of exposure. The STAT1 inhibitor epigallocatechin gallate (EGCG) attenuated STAT1 phosphorylation in cisplatin-treated utricles and resulted in concentration-dependent increases in hair cell survival at 24 h postexposure. Furthermore, we show that utricular hair cells from STAT1-deficient mice are resistant to cisplatin toxicity. EGCG failed to provide additional protection from cisplatin in STAT1-deficient mice, further supporting the hypothesis that the protective effects of EGCG are due to its inhibition of STAT1. Treatment with IFN-gamma, which also causes STAT1 activation, also induced hair cell death in wild-type but not STAT1-deficient mice. These results show that STAT1 is required for maximal cisplatin-induced hair cell death in the mouse utricle and suggest that treatment with EGCG may be a useful strategy for prevention of cisplatin ototoxicity.
Collapse
Affiliation(s)
- Nicole C. Schmitt
- Virginia Merrill Bloedel Hearing Research Center
- Department of Otolaryngology, Head and Neck Surgery, and
| | - Edwin W Rubel
- Virginia Merrill Bloedel Hearing Research Center
- Department of Otolaryngology, Head and Neck Surgery, and
| | - Neil M. Nathanson
- Virginia Merrill Bloedel Hearing Research Center
- Department of Pharmacology, University of Washington, Seattle, Washington 98195
| |
Collapse
|
49
|
Shaw PJ, Ditewig AC, Waring JF, Liguori MJ, Blomme EA, Ganey PE, Roth RA. Coexposure of mice to trovafloxacin and lipopolysaccharide, a model of idiosyncratic hepatotoxicity, results in a unique gene expression profile and interferon gamma-dependent liver injury. Toxicol Sci 2009; 107:270-80. [PMID: 18930950 PMCID: PMC2638649 DOI: 10.1093/toxsci/kfn205] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Accepted: 09/23/2008] [Indexed: 11/13/2022] Open
Abstract
The antibiotic trovafloxacin (TVX) has caused severe idiosyncratic hepatotoxicity in people, whereas levofloxacin (LVX) has not. Mice cotreated with TVX and lipopolysaccharide (LPS), but not with LVX and LPS, develop severe hepatocellular necrosis. Mice were treated with TVX and/or LPS, and hepatic gene expression changes were measured before liver injury using gene array. Hepatic gene expression profiles from mice treated with TVX/LPS clustered differently from those treated with LPS or TVX alone. Several of the probe sets expressed differently in TVX/LPS-treated mice were involved in interferon (IFN) signaling and the janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. A time course of plasma concentrations of IFN-gamma and interleukin (IL)-18, which directly induces IFN-gamma production, revealed that both cytokines were selectively increased in TVX/LPS-treated mice. Both IL-18(-/-) and IFN-gamma(-/-) mice were significantly protected from TVX/LPS-induced liver injury. In addition, IFN-gamma(-/-) mice had decreased plasma concentrations of tumor necrosis factor-alpha, IL-18, and IL-1beta when compared to wild-type mice. In conclusion, the altered expression of genes involved in IFN signaling in TVX/LPS-treated mice led to the finding that IL-18 and IFN-gamma play a critical role in TVX/LPS-induced liver injury.
Collapse
Affiliation(s)
- Patrick J. Shaw
- Department of Pharmacology and Toxicology, National Food Safety and Toxicology Center, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Amy C. Ditewig
- Department of Cell and Molecular Toxicology, Abbott Laboratories, Abbott Park, Illinois 60064
| | - Jeffrey F. Waring
- Department of Cell and Molecular Toxicology, Abbott Laboratories, Abbott Park, Illinois 60064
| | - Michael J. Liguori
- Department of Cell and Molecular Toxicology, Abbott Laboratories, Abbott Park, Illinois 60064
| | - Eric A. Blomme
- Department of Cell and Molecular Toxicology, Abbott Laboratories, Abbott Park, Illinois 60064
| | - Patricia E. Ganey
- Department of Pharmacology and Toxicology, National Food Safety and Toxicology Center, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Robert A. Roth
- Department of Pharmacology and Toxicology, National Food Safety and Toxicology Center, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
50
|
Hsieh PS, Chan JYH, Shyu JF, Chen YT, Loh CH. Mild portal endotoxaemia induces subacute hepatic inflammation and pancreatic beta-cell dysfunction in rats. Eur J Clin Invest 2008; 38:640-8. [PMID: 18837740 DOI: 10.1111/j.1365-2362.2008.01991.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Portal endotoxaemia has been speculated to be crucially involved in the pathogenesis of chronic hepatic inflammation, which is highly associated with the development of type 2 diabetes mellitus. This study tests whether portal endotoxaemia is a pathogenic link between chronic subacute hepatic inflammation and pancreatic beta-cell dysfunction. MATERIALS AND METHODS Rats were randomly assigned into two groups: rats with intraportal saline or low-dose lipopolysaccharide (LPS) infusion for 4 weeks. Pathological changes in the liver were evaluated via histological and biochemical examination. Pancreatic insulin secretion was evaluated by in vivo hyperglycaemic clamp study. RESULTS White blood cell count was significantly increased after intraportal LPS infusion for 4 weeks. Plasma amylase and chemoluminescence counts indicating superoxide levels were significantly increased after LPS treatments for 2 and 4 weeks. Intraportal low-dose LPS infusion significantly increased tumour necrosis factor-alpha and interleukin-6 contents in liver and pancreas. Circulating C-reactive protein, thiobarbituric acid reactive substances (TBARS) and endotoxin levels were not different among groups. The first- and second-phase insulin secretions in hyperglycaemic clamp were significantly decreased in LPS-treated rats. The histopathological scores, de novo production of reactive oxygen substrate and TBARS contents in the liver and pancreas were significantly increased in LPS-infused rats. Leucocyte infiltration was clearly visible in pancreatic islets of LPS-treated rats. CONCLUSIONS The present study demonstrated that mild portal endotoxaemia caused subacute hepatic inflammation and impaired pancreatic insulin secretion, implicating that portal endotoxaemia is a potential risk factor to link chronic subacute hepatic inflammation and pancreatic beta-cell dysfunction.
Collapse
Affiliation(s)
- P-S Hsieh
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan.
| | | | | | | | | |
Collapse
|