151
|
Qin M, Liu Y, Sun M, Li X, Xu J, Zhang L, Jiang H. Protective effects of melatonin on the white matter damage of neonatal rats by regulating NLRP3 inflammasome activity. Neuroreport 2021; 32:739-747. [PMID: 33994520 DOI: 10.1097/wnr.0000000000001642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of the study was to investigate the protective effects and relevant mechanisms of melatonin on the white matter damage (WMD) caused by endotoxin and ischemic hypoxia in neonatal rats. METHODS Seventy-two female neonatal rats (postnatal day 3) were randomly divided into the sham, melatonin-treated, and control groups (n = 24 for each group). The periventricular white matter was collected to evaluate the WMD and apoptosis. In addition, the reactive oxygen species (ROS) level was measured. The expression levels of nucleotide-binding domain-like receptor protein 3 (NLRP3), interleukin (IL)-1β, IL-18, pink1, parkin, Toll-like receptor (TLR)-4, and nuclear factor (NF)-κB were detected. RESULTS Hematoxylin and eosin and terminal-deoxynucleoitidyl transferase mediated nick end labeling staining showed that the WMD, as well as cell degeneration, necrosis, and apoptosis in the control group, were more severe than those in the melatonin-treated group. Endotoxin and ischemic hypoxia upregulated the expression of NLRP3 and downstream inflammatory factors such as IL-1β and IL-18, which could be reversed by melatonin treatment. Melatonin increased mitochondrial autophagy marker (pink1 and parkin) expression in the white matter and reduced ROS production. Moreover, melatonin-reduced TLR4 and NF-κB expression. CONCLUSIONS Melatonin can inhibit the hyperactivity of NLRP3 inflammasomes by enhancing mitochondrial autophagy and inhibiting TLR4/NF-κB pathway activity. Thus, melatonin may be a promising treatment for alleviating the WMD caused by endotoxin and ischemic hypoxia in neonatal rats.
Collapse
Affiliation(s)
- Miao Qin
- Department of Neonatology, Affiliated Hospital of Qingdao University, Qingdao, China
| | | | | | | | | | | | | |
Collapse
|
152
|
Belliere J, Casemayou A, Colliou E, El Hachem H, Kounde C, Piedrafita A, Feuillet G, Schanstra JP, Faguer S. Ibrutinib does not prevent kidney fibrosis following acute and chronic injury. Sci Rep 2021; 11:11985. [PMID: 34099830 PMCID: PMC8184891 DOI: 10.1038/s41598-021-91491-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/25/2021] [Indexed: 01/28/2023] Open
Abstract
Recent studies suggested that ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor, developed for the treatment of chronic lymphocytic leukemia, may prevent NLRP3 inflammasome activation in macrophages, IL-1β secretion and subsequent development of inflammation and organ fibrosis. The role of NLRP3 has been underlined in the various causes of acute kidney injury (AKI), a pathology characterized by high morbimortality and risk of transition toward chronic kidney disease (CKD). We therefore hypothesized that the BTK-inhibitor ibrutinib could be a candidate drug for AKI treatment. Here, we observed in both an AKI model (glycerol-induced rhabdomyolysis) and a model of rapidly progressive kidney fibrosis (unilateral ureteral obstruction), that ibrutinib did not prevent inflammatory cell recruitment in the kidney and fibrosis. Moreover, ibrutinib pre-exposure led to high mortality rate owing to severer rhabdomyolysis and AKI. In vitro, ibrutinib potentiated or had no effect on the secretion of IL-1β by monocytes exposed to uromodulin or myoglobin, two danger-associated molecule patterns proteins involved in the AKI to CKD transition. According to these results, ibrutinib should not be considered a candidate drug for patients developing AKI.
Collapse
Affiliation(s)
- Julie Belliere
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Audrey Casemayou
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Eloïse Colliou
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Hélène El Hachem
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Clément Kounde
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Alexis Piedrafita
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Guylène Feuillet
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
| | - Joost P Schanstra
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
| | - Stanislas Faguer
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France.
- Université Paul Sabatier-Toulouse 3, Toulouse, France.
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France.
| |
Collapse
|
153
|
Péladeau C, Sandhu JK. Aberrant NLRP3 Inflammasome Activation Ignites the Fire of Inflammation in Neuromuscular Diseases. Int J Mol Sci 2021; 22:ijms22116068. [PMID: 34199845 PMCID: PMC8200055 DOI: 10.3390/ijms22116068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are molecular hubs that are assembled and activated by a host in response to various microbial and non-microbial stimuli and play a pivotal role in maintaining tissue homeostasis. The NLRP3 is a highly promiscuous inflammasome that is activated by a wide variety of sterile triggers, including misfolded protein aggregates, and drives chronic inflammation via caspase-1-mediated proteolytic cleavage and secretion of proinflammatory cytokines, interleukin-1β and interleukin-18. These cytokines further amplify inflammatory responses by activating various signaling cascades, leading to the recruitment of immune cells and overproduction of proinflammatory cytokines and chemokines, resulting in a vicious cycle of chronic inflammation and tissue damage. Neuromuscular diseases are a heterogeneous group of muscle disorders that involve injury or dysfunction of peripheral nerves, neuromuscular junctions and muscles. A growing body of evidence suggests that dysregulation, impairment or aberrant NLRP3 inflammasome signaling leads to the initiation and exacerbation of pathological processes associated with neuromuscular diseases. In this review, we summarize the available knowledge about the NLRP3 inflammasome in neuromuscular diseases that affect the peripheral nervous system and amyotrophic lateral sclerosis, which affects the central nervous system. In addition, we also examine whether therapeutic targeting of the NLRP3 inflammasome components is a viable approach to alleviating the detrimental phenotype of neuromuscular diseases and improving clinical outcomes.
Collapse
Affiliation(s)
- Christine Péladeau
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada;
| | - Jagdeep K. Sandhu
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Correspondence: ; Tel.: +1-613-993-5304
| |
Collapse
|
154
|
Neys SFH, Hendriks RW, Corneth OBJ. Targeting Bruton's Tyrosine Kinase in Inflammatory and Autoimmune Pathologies. Front Cell Dev Biol 2021; 9:668131. [PMID: 34150760 PMCID: PMC8213343 DOI: 10.3389/fcell.2021.668131] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) was discovered due to its importance in B cell development, and it has a critical role in signal transduction downstream of the B cell receptor (BCR). Targeting of BTK with small molecule inhibitors has proven to be efficacious in several B cell malignancies. Interestingly, recent studies reveal increased BTK protein expression in circulating resting B cells of patients with systemic autoimmune disease (AID) compared with healthy controls. Moreover, BTK phosphorylation following BCR stimulation in vitro was enhanced. In addition to its role in BCR signaling, BTK is involved in many other pathways, including pattern recognition, Fc, and chemokine receptor signaling in B cells and myeloid cells. This broad involvement in several immunological pathways provides a rationale for the targeting of BTK in the context of inflammatory and systemic AID. Accordingly, numerous in vitro and in vivo preclinical studies support the potential of BTK targeting in these conditions. Efficacy of BTK inhibitors in various inflammatory and AID has been demonstrated or is currently evaluated in clinical trials. In addition, very recent reports suggest that BTK inhibition may be effective as immunosuppressive therapy to diminish pulmonary hyperinflammation in coronavirus disease 2019 (COVID-19). Here, we review BTK's function in key signaling pathways in B cells and myeloid cells. Further, we discuss recent advances in targeting BTK in inflammatory and autoimmune pathologies.
Collapse
|
155
|
Bai R, Lang Y, Shao J, Deng Y, Refuhati R, Cui L. The Role of NLRP3 Inflammasome in Cerebrovascular Diseases Pathology and Possible Therapeutic Targets. ASN Neuro 2021; 13:17590914211018100. [PMID: 34053242 PMCID: PMC8168029 DOI: 10.1177/17590914211018100] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cerebrovascular diseases are pathological conditions involving impaired blood flow in the brain, primarily including ischaemic stroke, intracranial haemorrhage, and subarachnoid haemorrhage. The nucleotide-binding and oligomerisation (NOD) domain-like receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3) inflammasome is a protein complex and a vital component of the immune system. Emerging evidence has indicated that the NLRP3 inflammasome plays an important role in cerebrovascular diseases. The function of the NLRP3 inflammasome in the pathogenesis of cerebrovascular diseases remains an interesting field of research. In this review, we first summarised the pathological mechanism of cerebrovascular diseases and the pathological mechanism of the NLRP3 inflammasome in aggravating atherosclerosis and cerebrovascular diseases. Second, we outlined signalling pathways through which the NLRP3 inflammasome participates in aggravating or mitigating cerebrovascular diseases. Reactive oxygen species (ROS)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), ROS/thioredoxin-interacting protein (TXNIP) and purinergic receptor-7 (P2X7R) signalling pathways can activate the NLRP3 inflammasome; activation of the NLRP3 inflammasome can aggravate cerebrovascular diseases by mediating apoptosis and pyroptosis. Autophagy/mitochondrial autophagy, nuclear factor E2-related factor-2 (Nrf2), interferon (IFN)-β, sirtuin (SIRT), and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) reportedly alleviate cerebrovascular diseases by inhibiting NLRP3 inflammasome activation. Finally, we explored specific inhibitors of the NLRP3 inflammasome based on the two-step activation of the NLRP3 inflammasome, which can be developed as new drugs to treat cerebrovascular diseases.
Collapse
Affiliation(s)
- Rongrong Bai
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yue Lang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Shao
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Deng
- Department of Hepatopancreatobiliary Surgery, The First Hospital of Jilin University, Changchun, China
| | - Reyisha Refuhati
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
156
|
Hibino S, Kawazoe T, Kasahara H, Itoh S, Ishimoto T, Sakata-Yanagimoto M, Taniguchi K. Inflammation-Induced Tumorigenesis and Metastasis. Int J Mol Sci 2021; 22:ijms22115421. [PMID: 34063828 PMCID: PMC8196678 DOI: 10.3390/ijms22115421] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation, especially chronic inflammation, plays a pivotal role in tumorigenesis and metastasis through various mechanisms and is now recognized as a hallmark of cancer and an attractive therapeutic target in cancer. In this review, we discuss recent advances in molecular mechanisms of how inflammation promotes tumorigenesis and metastasis and suppresses anti-tumor immunity in various types of solid tumors, including esophageal, gastric, colorectal, liver, and pancreatic cancer as well as hematopoietic malignancies.
Collapse
Affiliation(s)
- Sana Hibino
- Research Center for Advanced Science and Technology, Department of Inflammology, The University of Tokyo, Tokyo 153-0041, Japan;
| | - Tetsuro Kawazoe
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Hidenori Kasahara
- National Center for Global Health and Medicine, Department of Stem Cell Biology, Research Institute, Tokyo 162-8655, Japan;
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Takatsugu Ishimoto
- Gastrointestinal Cancer Biology, International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto 860-0811, Japan;
| | | | - Koji Taniguchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
- Correspondence: ; Tel.: +81-11-706-5050
| |
Collapse
|
157
|
Bi F, Zhang Y, Liu W, Xie K. Sinomenine activation of Nrf2 signaling prevents inflammation and cerebral injury in a mouse model of ischemic stroke. Exp Ther Med 2021; 21:647. [PMID: 33968178 PMCID: PMC8097210 DOI: 10.3892/etm.2021.10079] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Sinomenine (SINO), which is used clinically to treat rheumatoid arthritis and neuralgia, is derived from the root and stems of Sinomenium acutum. SINO has been reported to exert analgesic, sedative and anti-inflammatory effects, and provides a protective role against shock and organ damage. Studies have suggested that SINO primarily exerts it anti-inflammatory function by inhibiting NF-κB signaling. There is also evidence to indicate that SINO may regulate inflammation Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling. The present study aimed to investigate whether the anti-inflammatory and cerebral protective effects of SINO were induced through Nrf2 both in vitro and in vivo. The results revealed that SINO significantly upregulated Nrf2 protein expression levels, increased Nrf2 nuclear translocation and the upregulated the protein expression levels of downstream factors. The treatment of a middle cerebral artery occlusion model mice with SINO effectively reduced cerebral damage and inflammation, and restored the balance in cerebral oxidative stress. In addition, SINO treatment also promoted Nrf2-dependent microglia M1/M2 polarization and inhibited the phosphorylation of IκBα as well as NF-κB nuclear translocation. This revealed an important upstream event that contributed to its anti-inflammatory and cerebral tissue protective effects. In conclusion, the findings of the present study identified a novel pathway through which SINO may exert its anti-inflammatory and cerebral protective functions, and provided a molecular basis for the potential applications of SINO in the treatment of cerebral inflammatory disorders.
Collapse
Affiliation(s)
- Fangfang Bi
- Department of Medicine, Xi'an Peihua University, Xi'an, Shaanxi 710125, P.R. China
| | - Yiyong Zhang
- Department of Neurosurgery, Jinan Jiyang District People's Hospital, Jinan, Shandong 251401, P.R. China
| | - Wenbo Liu
- Department of Intensive Care Medicine, College of Anesthesiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Keliang Xie
- Department of Neurosurgery, Jinan Jiyang District People's Hospital, Jinan, Shandong 251401, P.R. China.,Department of Anesthesiology, First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, Shandong 261000, P.R. China
| |
Collapse
|
158
|
Li Y, Zhao J, Wu Y, Xia L. Btk knockout attenuates the liver inflammation in STZ-induced diabetic mice by suppressing NLRP3 inflammasome activation. Biochem Biophys Res Commun 2021; 549:75-82. [PMID: 33667712 DOI: 10.1016/j.bbrc.2021.02.094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 02/08/2023]
Abstract
Btk has pro-inflammatory role through a variety of signaling pathways. NLRP3 inflammasome plays a central role in liver inflammation for mediating the secretion of pro-inflammatory mediators. However, it is still unknown whether Btk could regulate NLRP3 inflammasome activation in diabetic liver. In this study, we used Btk knockout mice to establish the diabetic model by STZ. We found that Btk knockout could alleviate diabetic liver injury. This protection was due to reduced liver inflammation rather than lipid metabolism. Moreover, we found that macrophage infiltration and pro-inflammatory mediators were both significantly increased in diabetic mice liver. However, Btk deletion could reduce the activation of macrophage and secretion of pro-inflammatory cytokine, and reduced the liver inflammation through suppressing NLRP3 inflammasome activation. In conclusion, our study demonstrated that Btk knockout could significantly attenuate liver inflammation in diabetic mice by down-regulating NLRP3 inflammasome activation. Our finding has a broad prospect and provide a new idea for the treatment of diabetic liver injury.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Zhao
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yonggui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Lingling Xia
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
159
|
An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cell Mol Immunol 2021; 18:1141-1160. [PMID: 33850310 PMCID: PMC8093260 DOI: 10.1038/s41423-021-00670-3] [Citation(s) in RCA: 405] [Impact Index Per Article: 101.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/25/2021] [Indexed: 02/08/2023] Open
Abstract
The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a multiprotein complex involved in the release of mature interleukin-1β and triggering of pyroptosis, which is of paramount importance in a variety of physiological and pathological conditions. Over the past decade, considerable advances have been made in elucidating the molecular mechanisms underlying the priming/licensing (Signal 1) and assembly (Signal 2) involved in NLRP3 inflammasome activation. Recently, a number of studies have indicated that the priming/licensing step is regulated by complicated mechanisms at both the transcriptional and posttranslational levels. In this review, we discuss the current understanding of the mechanistic details of NLRP3 inflammasome activation with a particular emphasis on protein-protein interactions, posttranslational modifications, and spatiotemporal regulation of the NLRP3 inflammasome machinery. We also present a detailed summary of multiple positive and/or negative regulatory pathways providing upstream signals that culminate in NLRP3 inflammasome complex assembly. A better understanding of the molecular mechanisms underlying NLRP3 inflammasome activation will provide opportunities for the development of methods for the prevention and treatment of NLRP3 inflammasome-related diseases.
Collapse
|
160
|
Good L, Benner B, Carson WE. Bruton's tyrosine kinase: an emerging targeted therapy in myeloid cells within the tumor microenvironment. Cancer Immunol Immunother 2021; 70:2439-2451. [PMID: 33818636 PMCID: PMC8019691 DOI: 10.1007/s00262-021-02908-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022]
Abstract
Bruton’s tyrosine kinase (BTK) is a non-receptor kinase belonging to the Tec family of kinases. The role of BTK in B cell receptor signaling is well defined and is known to play a key role in the proliferation and survival of malignant B cells. Moreover, BTK has been found to be expressed in cells of the myeloid lineage. BTK has been shown to contribute to a variety of cellular pathways in myeloid cells including signaling in the NLRP3 inflammasome, receptor activation of nuclear factor-κβ and inflammation, chemokine receptor activation affecting migration, and phagocytosis. Myeloid cells are crucial components of the tumor microenvironment and suppressive myeloid cells contribute to cancer progression, highlighting a potential role for BTK inhibition in the treatment of malignancy. The increased interest in BTK inhibition in cancer has resulted in many preclinical studies that are testing the efficacy of using single-agent BTK inhibitors. Moreover, the ability of tumor cells to develop resistance to single-agent checkpoint inhibitors has resulted in clinical studies utilizing BTK inhibitors in combination with these agents to improve clinical responses. Furthermore, BTK regulates the immune response in microbial and viral infections through B cells and myeloid cells such as monocytes and macrophages. In this review, we describe the role that BTK plays in supporting suppressive myeloid cells, including myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM), while also discussing the anticancer effects of BTK inhibition and briefly describe the role of BTK signaling and BTK inhibition in microbial and viral infections.
Collapse
Affiliation(s)
- Logan Good
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Brooke Benner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - William E Carson
- Department of Surgery, Division of Surgical Oncology, Tzagournis Medical Research Facility, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
161
|
Vogel S, Kamimura S, Arora T, Smith ML, Almeida LEF, Combs CA, Thein SL, Quezado ZMN. NLRP3 inflammasome and bruton tyrosine kinase inhibition interferes with upregulated platelet aggregation and in vitro thrombus formation in sickle cell mice. Biochem Biophys Res Commun 2021; 555:196-201. [PMID: 33831782 DOI: 10.1016/j.bbrc.2021.03.115] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/21/2021] [Indexed: 12/20/2022]
Abstract
The nucleotide-binding domain leucine-rich repeat containing protein 3 (NLRP3) inflammasome is a critical inflammatory mechanism identified in platelets, which controls platelet activation and aggregation. We have recently shown that the platelet NLRP3 inflammasome is upregulated in sickle cell disease (SCD), which is mediated by Bruton tyrosine kinase (BTK). Here, we investigated the effect of pharmacological inhibition of NLRP3 and BTK on platelet aggregation and the formation of in vitro thrombi in Townes SCD mice. Mice were injected for 4 weeks with the NLRP3 inhibitor MCC950, the BTK inhibitor ibrutinib or vehicle control. NLRP3 activity, as monitored by caspase-1 activation, was upregulated in platelets from SCD mice, which was dependent on BTK. Large areas of platelet aggregates detected in the liver of SCD mice were decreased when mice were treated with MCC950 or ibrutinib. Moreover, platelet aggregation and in vitro thrombus formation were upregulated in SCD mice and were inhibited when mice were subjected to pharmacological inhibition of NLRP3 and BTK. Targeting the NLRP3 inflammasome might be a novel approach for antiplatelet therapy in SCD.
Collapse
Affiliation(s)
- Sebastian Vogel
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Sayuri Kamimura
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Taruna Arora
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meghann L Smith
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Luis E F Almeida
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Christian A Combs
- Light Microscopy Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zenaide M N Quezado
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA; Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
162
|
Zhang Y, Wang H, Li H, Nan L, Xu W, Lin Y, Chu K. Gualou Guizhi Granule Protects against OGD/R-Induced Injury by Inhibiting Cell Pyroptosis via the PI3K/Akt Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6613572. [PMID: 33747105 PMCID: PMC7960020 DOI: 10.1155/2021/6613572] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/05/2021] [Accepted: 02/28/2021] [Indexed: 01/30/2023]
Abstract
Pyroptosis is a proinflammatory form of regulated cell death that plays an important role in ischemic stroke. Gualou Guizhi granule (GLGZG) is a classic prescription that has been shown to exert neuroprotective effects against cerebral ischemia reperfusion injury. In the present study, we examined the involvement of pyroptosis and its associated mechanism in protecting nerve function. Methods. Primary neurons were exposed to oxygen-glucose deprivation and reperfusion (OGD/R) conditions in the presence or absence of GLGZG. Cellular viability was measured by the 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazoliumbromide (MTT) assay. The number of apoptoic cells was detected by NeuN and NSE protein expression. The expression levels of the pyroptosis markers, namely, NOD-like receptor family pyrin domain-containing 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, interleukin-18 (IL-18), and IL-1β were determined by quantitative real-time PCR analysis, western blot, and ELISA analyses as appropriate. Moreover, the expression levels of the PI3K/Akt pathway key proteins were determined by quantitative real-time PCR analysis and western blot assays. To determine the PI3K/Akt pathway involvement in GLGZG-mediated neuroprotection, the PI3K inhibitor LY294002 (LY, 10 μM) was added. The expression levels of NeuN, Akt, and p-Akt were evaluated. Results. It was found that GLGZG could inhibit OGD/R-induced cell apoptosis, increase neuronal cell viability, decrease the production of IL-18 and IL-1β, and downregulate the expression levels of pyroptosis markers (NLRP3, ASC, and caspase-1). Furthermore, GLGZG could modulate the PI3K/Akt signaling pathway. Pharmacological inhibition of the PI3K pathway not only abrogated the effects of GLGZG on Akt but also neutralized its prosurvival and antipyroptotic actions. Conclusions. The findings indicated that GLGZG pretreatment effectively reduced OGD/R-induced injury by inhibiting cell pyroptosis and activating the PI3K/Akt pathway. These data provide important evidence for the therapeutic applications of this regimen in ischemic stroke.
Collapse
Affiliation(s)
- Yuqin Zhang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- State Key Laboratory of Chinese Pharmacies, Fujian Provincial Department of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Hongyun Wang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- State Key Laboratory of Chinese Pharmacies, Fujian Provincial Department of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Huang Li
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- State Key Laboratory of Chinese Pharmacies, Fujian Provincial Department of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Lihong Nan
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- State Key Laboratory of Chinese Pharmacies, Fujian Provincial Department of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Wei Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- State Key Laboratory of Chinese Pharmacies, Fujian Provincial Department of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yu Lin
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- State Key Laboratory of Chinese Pharmacies, Fujian Provincial Department of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Kedan Chu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- State Key Laboratory of Chinese Pharmacies, Fujian Provincial Department of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| |
Collapse
|
163
|
Lee H, Jeon SG, Kim J, Kang RJ, Kim S, Han K, Park H, Kim K, Sung YM, Nam HY, Koh YH, Song M, Suk K, Hoe H. Ibrutinib modulates Aβ/tau pathology, neuroinflammation, and cognitive function in mouse models of Alzheimer's disease. Aging Cell 2021; 20:e13332. [PMID: 33709472 PMCID: PMC7963331 DOI: 10.1111/acel.13332] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 12/23/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
We previously demonstrated that ibrutinib modulates LPS‐induced neuroinflammation in vitro and in vivo, but its effects on the pathology of Alzheimer's disease (AD) and cognitive function have not been investigated. Here, we investigated the effects of ibrutinib in two mouse models of AD. In 5xFAD mice, ibrutinib injection significantly reduced Aβ plaque levels by promoting the non‐amyloidogenic pathway of APP cleavage, decreased Aβ‐induced neuroinflammatory responses, and significantly downregulated phosphorylation of tau by reducing levels of phosphorylated cyclin‐dependent kinase‐5 (p‐CDK5). Importantly, tau‐mediated neuroinflammation and tau phosphorylation were also alleviated by ibrutinib injection in PS19 mice. In 5xFAD mice, ibrutinib improved long‐term memory and dendritic spine number, whereas in PS19 mice, ibrutinib did not alter short‐ and long‐term memory but promoted dendritic spinogenesis. Interestingly, the induction of dendritic spinogenesis by ibrutinib was dependent on the phosphorylation of phosphoinositide 3‐kinase (PI3K). Overall, our results suggest that ibrutinib modulates AD‐associated pathology and cognitive function and may be a potential therapy for AD.
Collapse
Affiliation(s)
- Hyun‐ju Lee
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
| | - Seong Gak Jeon
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
| | - Jieun Kim
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
| | - Ri Jin Kang
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
| | - Seong‐Min Kim
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
- Medical Device Development Center Daegu‐Gyeongbuk Medical Innovation Foundation (DGMIF) Daegu Korea
| | - Kyung‐Min Han
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
| | - HyunHee Park
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
| | - Ki‐taek Kim
- Department of Life Sciences Yeungnam University Gyeongsan Korea
| | - You Me Sung
- Korea Mouse Phenotyping Center (KMPC) Seoul National University Seoul Korea
| | - Hye Yeon Nam
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
| | - Young Ho Koh
- Center for Biomedical Sciences Center for Infectious Diseases Division of Brain Disease Korea National Institute of Health Heungdeok‐gu Korea
| | - Minseok Song
- Department of Life Sciences Yeungnam University Gyeongsan Korea
| | - Kyoungho Suk
- Department of Pharmacology Brain Science & Engineering Institute School of Medicine Kyungpook National University Daegu Korea
| | - Hyang‐Sook Hoe
- Department of Neural Development and Disease Korea Brain Research Institute (KBRI) Daegu Korea
- Department of Brain and Cognitive Sciences Daegu Gyeongbuk Institute of Science & Technology Daegu Korea
| |
Collapse
|
164
|
Weber ANR. Targeting the NLRP3 Inflammasome via BTK. Front Cell Dev Biol 2021; 9:630479. [PMID: 33718366 PMCID: PMC7947255 DOI: 10.3389/fcell.2021.630479] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/14/2021] [Indexed: 12/28/2022] Open
Abstract
The NLRP3 inflammasome represents a critical inflammatory machinery driving pathology in many acute (e. g., myocardial infarction or stroke) and chronic (Alzheimer's disease, atherosclerosis) human disorders linked to the activity of IL-1 cytokines. Although the therapeutic potential of NLRP3 is undisputed, currently no clinically approved therapies exist to target the NLRP3 inflammasome directly. The recent discovery of BTK as a direct and positive regulator of the NLRP3 inflammasome has, however, raised the intriguing possibility of targeting the NLRP3 inflammasome via existing or future BTK inhibitors. Here, I review the mechanistic basis for this notion and discuss the molecular and cellular role of BTK in the inflammasome process. Specific attention will be given to cell-type dependent characteristics and differences that may be relevant for targeting approaches. Furthermore, I review recent (pre-)clinical evidence for effects of BTK inhibitors on NLRP3 activity and highlight and discuss open questions and future research directions. Collectively, the concept of targeting BTK to target NLRP3-dependent inflammation will be explored comprehensively at the molecular, cellular and therapeutic levels.
Collapse
Affiliation(s)
- Alexander N. R. Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- iFIT – Cluster of Excellence (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- CMFI – Cluster of Excellence (EXC 2124) “Controlling Microbes to Fight Infection”, University of Tübingen, Tübingen, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK; German Cancer Consortium), Partner Site Tübingen, Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
165
|
Liang Z, Damianou A, Di Daniel E, Kessler BM. Inflammasome activation controlled by the interplay between post-translational modifications: emerging drug target opportunities. Cell Commun Signal 2021; 19:23. [PMID: 33627128 PMCID: PMC7905589 DOI: 10.1186/s12964-020-00688-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/23/2020] [Indexed: 12/27/2022] Open
Abstract
Controlling the activation of the NLRP3 inflammasome by post-translational modifications (PTMs) of critical protein subunits has emerged as a key determinant in inflammatory processes as well as in pathophysiology. In this review, we put into context the kinases, ubiquitin processing and other PTM enzymes that modify NLRP3, ASC/PYCARD and caspase-1, leading to inflammasome regulation, activation and signal termination. Potential target therapeutic entry points for a number of inflammatory diseases focussed on PTM enzyme readers, writers and erasers, leading to the regulation of inflammasome function, are discussed. Video Abstract.
Collapse
Affiliation(s)
- Zhu Liang
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ UK
- Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ UK
| | - Andreas Damianou
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ UK
| | - Elena Di Daniel
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ UK
- ARUK Oxford Drug Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ UK
| | - Benedikt M. Kessler
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ UK
- Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ UK
| |
Collapse
|
166
|
Riaz M, Rehman AU, Shah SA, Rafiq H, Lu S, Qiu Y, Wadood A. Predicting Multi-Interfacial Binding Mechanisms of NLRP3 and ASC Pyrin Domains in Inflammasome Activation. ACS Chem Neurosci 2021; 12:603-612. [PMID: 33504150 DOI: 10.1021/acschemneuro.0c00519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
NLRP3-PYD inflammasome activates an inflammatory pathway in response to a wide variety of cell damage or infections. Dysregulated NLRP3 inflammatory signaling has many chronic inflammatory and autoimmune disorders. NLRP3 and ASC have a PYD, a superfamily member of the Death Domain, which plays a key role in inflammatory assembly. The ASC interacts with NLRP3 through a homotypic PYD and recruits the procaspase-1 through a homotypic caspase recruitment domain interaction. Here, we used several computational approaches to reveal the interactions of the NLRP3 and ASC PYD domains that lead to the activation of the inflammasome complex. We have characterized ASC and NLRP3-PYD intermolecular interactions by protein-protein docking, and further molecular dynamics (MD) simulations were conducted to evaluate the stability of NLRP3/ASC-PYD complex. Subsequently, we have identified several residues that stabilize the NLRP3/ASC-PYD complex in different faces (i.e., Face-1 to Face-4). The research framework offers new insights into the molecular mechanisms of inflammasome and apoptosis signaling as well as the ease of the drug discovery process.
Collapse
Affiliation(s)
- Muhammad Riaz
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Ashfaq Ur Rehman
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Shahid Ali Shah
- Department of Chemistry, Sarhad University of Science and Information Technology, Peshawar 25000, Pakistan
| | - Humaira Rafiq
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Yingying Qiu
- Department of Neurology, Tiantai Hospital of Traditional Chinese Medicine, Taizhou, Zhejiang 317200, China
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
167
|
Su M, Wang W, Liu F, Li H. Recent Progress on the Discovery of NLRP3 Inhibitors and their Therapeutic Potential. Curr Med Chem 2021; 28:569-582. [PMID: 31971103 DOI: 10.2174/0929867327666200123093544] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/18/2019] [Accepted: 11/24/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Inflammation is the body's immune system's fast coordinating response to irritants caused by pathogens, external injuries, and chemical or radiation effects. The nucleotidebinding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is a critical component of the innate immune system. The dysfunction of NLRP3 inflammasome contributes to various pathogeneses of complex diseases, such as uncontrolled infection, autoimmune diseases, neurodegenerative diseases, and metabolic disorders. This review describes recent progress on the discovery of NLRP3 inflammasome inhibitors and their therapeutic potential. METHODS Based on the mechanism of NLRP3 activation, several types of NLRP3 inhibitors are described and summarized according to their origins, structures, bioactivity, and mechanism of action. Structure-Activity Relationship (SAR) is also listed for different scaffolds, as well as effective pharmacophore. RESULTS Over one-hundred papers were included in the review. The development of NLRP3 inhibitors has been described from the earliest glyburide in 2001 to the latest progress in 2019. Several series of inhibitors have been categorized, such as JC-series based on glyburide and BC-series based on 2APB. Many other small molecules such as NLRP3 inhibitors are also listed. SAR, application in related therapeutic models, and five different action mechanisms are described. CONCLUSION The findings of this review confirmed the importance of developing NLRP3 inflammasome inhibitors. Various NLRP3 inhibitors have been discovered as effective therapeutic treatments for multiple diseases, such as type II diabetes, experimental autoimmune encephalomyelitis, stressrelated mood disorders, etc. The development of a full range of NLRP3 inflammasome inhibitors is still at its foundational phase. We are looking forward to the identification of inhibitory agents that provide the most potent therapeutic strategies and efficiently treat NLRP3 inflammasome-related inflammatory diseases.
Collapse
Affiliation(s)
- Ma Su
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, 1 Wenjing Road, Suzhou 215123, China
| | - Weiwei Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, 1 Wenjing Road, Suzhou 215123, China
| | - Feng Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, 1 Wenjing Road, Suzhou 215123, China
| | - Huanqiu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, 1 Wenjing Road, Suzhou 215123, China
| |
Collapse
|
168
|
Carinci M, Vezzani B, Patergnani S, Ludewig P, Lessmann K, Magnus T, Casetta I, Pugliatti M, Pinton P, Giorgi C. Different Roles of Mitochondria in Cell Death and Inflammation: Focusing on Mitochondrial Quality Control in Ischemic Stroke and Reperfusion. Biomedicines 2021; 9:biomedicines9020169. [PMID: 33572080 PMCID: PMC7914955 DOI: 10.3390/biomedicines9020169] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunctions are among the main hallmarks of several brain diseases, including ischemic stroke. An insufficient supply of oxygen and glucose in brain cells, primarily neurons, triggers a cascade of events in which mitochondria are the leading characters. Mitochondrial calcium overload, reactive oxygen species (ROS) overproduction, mitochondrial permeability transition pore (mPTP) opening, and damage-associated molecular pattern (DAMP) release place mitochondria in the center of an intricate series of chance interactions. Depending on the degree to which mitochondria are affected, they promote different pathways, ranging from inflammatory response pathways to cell death pathways. In this review, we will explore the principal mitochondrial molecular mechanisms compromised during ischemic and reperfusion injury, and we will delineate potential neuroprotective strategies targeting mitochondrial dysfunction and mitochondrial homeostasis.
Collapse
Affiliation(s)
- Marianna Carinci
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.C.); (B.V.); (S.P.); (P.P.)
| | - Bianca Vezzani
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.C.); (B.V.); (S.P.); (P.P.)
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.C.); (B.V.); (S.P.); (P.P.)
| | - Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany; (P.L.); (K.L.); (T.M.)
| | - Katrin Lessmann
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany; (P.L.); (K.L.); (T.M.)
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany; (P.L.); (K.L.); (T.M.)
| | - Ilaria Casetta
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (M.P.)
| | - Maura Pugliatti
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (M.P.)
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.C.); (B.V.); (S.P.); (P.P.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.C.); (B.V.); (S.P.); (P.P.)
- Correspondence:
| |
Collapse
|
169
|
Seok JK, Kang HC, Cho YY, Lee HS, Lee JY. Regulation of the NLRP3 Inflammasome by Post-Translational Modifications and Small Molecules. Front Immunol 2021; 11:618231. [PMID: 33603747 PMCID: PMC7884467 DOI: 10.3389/fimmu.2020.618231] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a host protection mechanism that eliminates invasive pathogens from the body. However, chronic inflammation, which occurs repeatedly and continuously over a long period, can directly damage tissues and cause various inflammatory and autoimmune diseases. Pattern recognition receptors (PRRs) respond to exogenous infectious agents called pathogen-associated molecular patterns and endogenous danger signals called danger-associated molecular patterns. Among PRRs, recent advancements in studies of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome have established its significant contribution to the pathology of various inflammatory diseases, including metabolic disorders, immune diseases, cardiovascular diseases, and cancer. The regulation of NLRP3 activation is now considered to be important for the development of potential therapeutic strategies. To this end, there is a need to elucidate the regulatory mechanism of NLRP3 inflammasome activation by multiple signaling pathways, post-translational modifications, and cellular organelles. In this review, we discuss the intracellular signaling events, post-translational modifications, small molecules, and phytochemicals participating in the regulation of NLRP3 inflammasome activation. Understanding how intracellular events and small molecule inhibitors regulate NLRP3 inflammasome activation will provide crucial information for elucidating the associated host defense mechanism and the development of efficient therapeutic strategies for chronic diseases.
Collapse
Affiliation(s)
- Jin Kyung Seok
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| | - Han Chang Kang
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| | - Yong-Yeon Cho
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| | - Hye Suk Lee
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| | - Joo Young Lee
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| |
Collapse
|
170
|
Franke M, Bieber M, Kraft P, Weber ANR, Stoll G, Schuhmann MK. The NLRP3 inflammasome drives inflammation in ischemia/reperfusion injury after transient middle cerebral artery occlusion in mice. Brain Behav Immun 2021; 92:223-233. [PMID: 33307174 DOI: 10.1016/j.bbi.2020.12.009] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/30/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Cerebral ischemia induces a profound neuro-inflammatory response, but the underlying molecular mechanisms are poorly understood. Inflammasomes (NLRP1, NLRP3, NLRC4, AIM2) are intracellular multi-protein complexes which can induce sets of pro-inflammatory cyto- and chemokines, and thereby guide inflammation. We, here, assessed the functional role of NLRP3 in ischemia/reperfusion (I/R) injury in a mouse model of transient cerebral ischemia. METHODS Ischemic stroke was induced in C57Bl/6 mice by 60 min transient middle cerebral artery occlusion (tMCAO) and 3, 7 or 23 h of reperfusion, a paradigm of I/R injury. The expression patterns of inflammasomes in the ischemic hemispheres were evaluated by semiquantitative real-time PCR and Western Blot analysis accompanied by protein localization using immunocytochemistry. Finally, animals were treated with the inflammasome inhibitors Sulforaphane, Genipin, MCC950 or vehicle, directly before or upon recanalization after tMCAO. Stroke outcome was assessed, including infarct size and functional deficits, local inflammatory response, neuronal survival as well as blood-brain barrier function on day 1 after tMCAO. RESULTS After tMCAO the relative gene expression levels of NLRP3 increased 20-30x within 1 day in the ischemic hemisphere which translated into an increased expression of NLRP3 in neurons. Accordingly, the gene expression levels of the NLRP3-modulator, Bruton's Tyrosine Kinase (BTK), and the NLRP3-inducible cytokine IL-1β significantly rose. Lesser or non-significant changes were seen for the other inflammasomes. Application of inflammasome inhibitors covering all inflammasomes or specifically NLRP3 significantly reduced infarct volumes when given before or after tMCAO and was accompanied by clear evidence for reduced activation of caspase 1. This stroke attenuating effect coincided with less immune cell infiltration in the ischemic hemisphere and preservation of the blood-brain barrier integrity. CONCLUSIONS Our data show that induction of the NLRP3 inflammasome in neurons drives neuroinflammation in acute ischemic stroke. Early blockade of NLRP3 protects from I/R injury by mitigating inflammation and stabilizing the blood-brain barrier.
Collapse
Affiliation(s)
- Maximilian Franke
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| | - Michael Bieber
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| | - Peter Kraft
- Department of Neurology, Klinikum Main-Spessart, Grafen-von-Rieneck-Str. 5, 97816 Lohr, Germany.
| | - Alexander N R Weber
- Interfaculty Institute of Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany; iFIT - Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Germany.
| | - Guido Stoll
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| | - Michael K Schuhmann
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| |
Collapse
|
171
|
Zou XS, Xie L, Wang WY, Zhao GY, Tian XY, Chen MH. Pomelo peel oil alleviates cerebral NLRP3 inflammasome activation in a cardiopulmonary resuscitation rat model. Exp Ther Med 2021; 21:233. [PMID: 33603841 PMCID: PMC7851623 DOI: 10.3892/etm.2021.9664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022] Open
Abstract
The NLR family pyrin domain-containing 3 (NLRP3) inflammasome, which is composed of NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC) and pro-caspase-1 protein complexes, is activated by the reactive oxygen species (ROS) that are associated with ischemia-reperfusion (I/R) and are involved in brain damage. Pomelo peel oil (PPO) exhibits antioxidant activity. However, it is unclear whether PPO is able to attenuate NLRP3 inflammasome-induced inflammation and pyroptosis. Healthy male Sprague-Dawley rats were subjected to 7 min of cardiac arrest via trans-esophageal electrical stimulation, followed by cardiopulmonary resuscitation (CPR). The rats were then treated with PPO prior to reperfusion for 24 h. Hematoxylin and eosin staining was used to evaluate brain tissue and cell damage. In the brain tissues, reactive oxygen species (ROS) were assayed, immunofluorescence was used to analyze the expression of NLRP3 and western blotting was performed to determine the expression levels of neuroenolase (NSE), NF-κB, interleukin-1β (IL-1β), gasdermin D (GSDMD) and the NLRP3 inflammasome. Treatment of the rats with PPO significantly decreased the pathological damage of the brain tissue and reduced the expression of NSE, production of ROS and secretion of NF-κB, NLRP3, IL-1β and GSDMD. In conclusion, these results demonstrate the ability of PPO to protect the brain against I/R injury in rats after CPR by a mechanism involving inhibition of the inflammation and pyroptosis mediated by NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xin-Sen Zou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Lu Xie
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Wen-Yan Wang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Gao-Yang Zhao
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Xin-Yue Tian
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Meng-Hua Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| |
Collapse
|
172
|
Benner B, Carson WE. Observations on the use of Bruton's tyrosine kinase inhibitors in SAR-CoV-2 and cancer. J Hematol Oncol 2021; 14:15. [PMID: 33441177 PMCID: PMC7805262 DOI: 10.1186/s13045-020-00999-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/12/2020] [Indexed: 01/05/2023] Open
Abstract
Bruton's tyrosine kinase (BTK) inhibitors, drugs utilized in cancer, are being repurposed for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) (COVID-19). Recently, BTK inhibitors acalabrutinib and ibrutinib have been found to protect against pulmonary injury in a small group of patients infected with SARS-CoV-2. The high levels of pro-inflammatory cytokines found in the circulation of COVID-19 patients with severe lung disease suggest the involvement of the innate immune system in this process. Understanding the potential mechanism of action of BTK inhibition in SARS-CoV-2 is clearly of importance to determine how acalabrutinib, ibrutinib and possibly other BTK inhibitors may provide protection against lung injury.
Collapse
Affiliation(s)
- Brooke Benner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - William E Carson
- Department of Surgery, Division of Surgical Oncology, The Ohio State University, N924 Doan Hall, 410 W. 10th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
173
|
Genetics and Pathogenetic Role of Inflammasomes in Philadelphia Negative Chronic Myeloproliferative Neoplasms: A Narrative Review. Int J Mol Sci 2021; 22:ijms22020561. [PMID: 33429941 PMCID: PMC7827003 DOI: 10.3390/ijms22020561] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/01/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The last decade has been very important for the quantity of preclinical information obtained regarding chronic myeloproliferative neoplasms (MPNs) and the following will be dedicated to the translational implications of the new biological acquisitions. The overcoming of the mechanistic model of clonal evolution and the entry of chronic inflammation and dysimmunity into the new model are the elements on which to base a part of future therapeutic strategies. The innate immune system plays a major role in this context. Protagonists of the initiation and regulation of many pathological aspects, from cytokine storms to fibrosis, the NLRP3 and AIM2 inflammasomes guide and condition the natural history of the disease. For this reason, MPNs share many biological and clinical aspects with non-neoplastic diseases, such as autoimmune disorders. Finally, cardiovascular risk and disturbances in iron metabolism and myelopoiesis are also closely linked to the role of inflammasomes. Although targeted therapies are already being tested, an increase in knowledge on the subject is desirable and potentially translates into better care for patients with MPNs.
Collapse
|
174
|
Wei J, Wang Y, Qi X, Wu Y. Enhanced Bruton's tyrosine kinase activity in the kidney of patients with IgA nephropathy. Int Urol Nephrol 2021; 53:1399-1415. [PMID: 33389462 PMCID: PMC8192408 DOI: 10.1007/s11255-020-02733-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022]
Abstract
Purpose Bruton’s tyrosine kinase (BTK) is a vital biological molecule that contributes to immune regulation. Previous studies have showed that BTK can be detected in patients with lupus nephritis and rheumatoid arthritis. However, the role of BTK in IgA nephropathy (IgAN) has not yet been elucidated. The purpose of this research was to investigate the role of BTK activation in macrophages in IgAN. Methods Peripheral blood and renal tissue samples were collected from 63 patients with IgAN, and peritumoral normal tissues were collected from 20 patients after surgical resection of renal tumor for use as control. Additionally, 20 healthy volunteers were recruited as control. The levels of BTK, CD68, phosphorylated BTK (pBTK), phosphorylated NF-κB (p-NF-κB p65), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and monocyte chemotactic protein (MCP)-1 were measured by immunohistochemistry (IHC), real-time polymerase chain reaction (RT-PCR), western blotting, and enzyme-linked immunosorbent assay (ELISA). Results Compared to peritumoral normal tissues, the expression levels of CD68 and BTK were significantly increased in IgAN group (p < 0.001) and the differences between M0 and M1, E0 and E1, S0 and S1, T0 and T1-2, C0 and C1-2 were statistically significant in the updated Oxford Classification (p < 0.05). Also, CD68 and BTK were positively correlated with Katafuchi semi-quantitative glomerular and tubulointerstitial scores (r = 0.580, 0.637 and 0.442, 0.489, respectively, p < 0.05). The expression of BTK was significantly higher in C3b- and C4d-positive renal tissues of patients with IgAN (p < 0.05). In addition, BTK was positively correlated with 24-h urine protein, serum creatinine levels (r = 0.456 and 0.453, respectively, p < 0.001), and negatively correlated with serum albumin (r = 0.357, p < 0.05). The intensity of expression of pBTK and p-NF-κB p65 was observably increased in renal tissues and monocytes of patients with IgAN compared to the control group. The results of IHC, RT-PCR, and ELISA indicated that the levels of TNF-ɑ, IL-1β, and MCP-1 were markedly increased in the IgAN group (p < 0.05). Conclusion The results of this study indicate that activation of BTK in macrophages may play an important role in promoting the progression of renal inflammation in IgAN.
Collapse
Affiliation(s)
- Jie Wei
- Department of Nephrology, The First Affiliated Hospital, Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230032, People's Republic of China
| | - Yan Wang
- Department of Nephrology, The First Affiliated Hospital, Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230032, People's Republic of China
| | - Xiangming Qi
- Department of Nephrology, The First Affiliated Hospital, Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230032, People's Republic of China
| | - Yonggui Wu
- Department of Nephrology, The First Affiliated Hospital, Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230032, People's Republic of China.
| |
Collapse
|
175
|
Hu X, Yan J, Huang L, Araujo C, Peng J, Gao L, Liu S, Tang J, Zuo G, Zhang JH. INT-777 attenuates NLRP3-ASC inflammasome-mediated neuroinflammation via TGR5/cAMP/PKA signaling pathway after subarachnoid hemorrhage in rats. Brain Behav Immun 2021; 91:587-600. [PMID: 32961266 PMCID: PMC7749833 DOI: 10.1016/j.bbi.2020.09.016] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Inflammasome-mediated neuroinflammation plays an important role in the pathogenesis of early brain injury (EBI) following subarachnoid hemorrhage (SAH). The activation of the TGR5 receptor has been shown to be neuroprotective in a variety of neurological diseases. This study aimed to investigate the effects of the specific synthetic TGR5 agonist, INT-777, in attenuating NLRP3-ASC inflammasome activation and reducing neuroinflammation after SAH. METHODS One hundred and eighty-four male Sprague Dawley rats were used. SAH was induced by the endovascular perforation. INT-777 was administered intranasally at 1 h after SAH induction. To elucidate the signaling pathway involved in the effect of INT-777 on inflammasome activation during EBI, TGR5 knockout CRISPR and PKA inhibitor H89 were administered intracerebroventricularly and intraperitoneally at 48 h and 1 h before SAH. The SAH grade, short- and long-term neurobehavioral assessments, brain water content, western blot, immunofluorescence staining, and Nissl staining were performed. RESULTS The expressions of endogenous TGR5, p-PKA, and NLRP3-ASC inflammasome were increased after SAH. INT-777 administration significantly decreased NLRP3-ASC inflammasome activation in microglia, reduced brain edema and neuroinflammation, leading to improved short-term neurobehavioral functions at 24 h after SAH. The administration of TGR5 CRISPR or PKA inhibitor (H89) abolished the anti-inflammation effects of INT-777, on NLRP3-ASC inflammasome, pro-inflammatory cytokines (IL-6, IL-1β, and TNF-a), and neutrophil infiltration at 24 h after SAH. Moreover, early administration of INT-777 attenuated neuronal degeneration in hippocampus on 28 d after SAH. CONCLUSIONS INT-777 attenuated NLRP3-ASC inflammasome-dependent neuroinflammation in the EBI after SAH, partially via TGR5/cAMP/PKA signaling pathway. Early administration of INT-777 may serve as a potential therapeutic strategy for EBI management in the setting of SAH.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jun Yan
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Camila Araujo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jun Peng
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570000, China
| | - Ling Gao
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570000, China
| | - Shengpeng Liu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Taicang Hospital Affiliated to Soochow University, Taicang, Suzhou, Jiangsu 215400, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA; Department of Anesthesiology, Loma Linda University, Loma Linda, CA 92350, USA.
| |
Collapse
|
176
|
Gustine JN, Jones D. Immunopathology of Hyperinflammation in COVID-19. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:4-17. [PMID: 32919977 PMCID: PMC7484812 DOI: 10.1016/j.ajpath.2020.08.009] [Citation(s) in RCA: 342] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 01/08/2023]
Abstract
The rapid spread of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), has resulted in an unprecedented public health crisis worldwide. Recent studies indicate that a hyperinflammatory syndrome induced by SARS-CoV-2 contributes to disease severity and mortality in COVID-19. In this review, an overview of the pathophysiology underlying the hyperinflammatory syndrome in severe COVID-19 is provided. The current evidence suggests that the hyperinflammatory syndrome results from a dysregulated host innate immune response. The gross and microscopic pathologic findings as well as the alterations in the cytokine milieu, macrophages/monocytes, natural killer cells, T cells, and neutrophils in severe COVID-19 are summarized. The data highlighted include the potential therapeutic approaches undergoing investigation to modulate the immune response and abrogate lung injury in severe COVID-19.
Collapse
Affiliation(s)
- Joshua N Gustine
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Dennis Jones
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
177
|
Lu LQ, Tian J, Luo XJ, Peng J. Targeting the pathways of regulated necrosis: a potential strategy for alleviation of cardio-cerebrovascular injury. Cell Mol Life Sci 2021; 78:63-78. [PMID: 32596778 PMCID: PMC11072340 DOI: 10.1007/s00018-020-03587-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/09/2020] [Accepted: 06/23/2020] [Indexed: 12/31/2022]
Abstract
Apoptosis, necrosis and autophagy-dependent cell death are the three major types of cell death. Traditionally, necrosis is thought as a passive and unregulated form of cell death. However, certain necrosis can also occur in a highly regulated manner, referring to regulated necrosis. Depending on the signaling pathways, regulated necrosis can be further classified as necroptosis, pyroptosis, ferroptosis, parthanatos and CypD-mediated necrosis. Numerous studies have reported that regulated necrosis contributes to the progression of multiple injury-relevant diseases. For example, necroptosis contributes to the development of myocardial infarction, atherosclerosis, heart failure and stroke; pyroptosis is involved in the progression of myocardial or cerebral infarction, atherosclerosis and diabetic cardiomyopathy; while ferroptosis, parthanatos and CypD-mediated necrosis participate in the pathological process of myocardial and/or cerebral ischemia/reperfusion injury. Thereby, targeting the pathways of regulated necrosis pharmacologically or genetically could be an efficient strategy for reducing cardio-cerebrovascular injury. Further study needs to focus on the crosstalk and interplay among different types of regulated necrosis. Pharmacological intervention of two or more types of regulated necrosis simultaneously may have advantages in clinic to treat injury-relevant diseases.
Collapse
Affiliation(s)
- Li-Qun Lu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jing Tian
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
178
|
Nadeem A, Ahmad SF, Al-Harbi NO, Ibrahim KE, Alqahtani F, Alanazi WA, Mahmood HM, Alsanea S, Attia SM. Bruton's tyrosine kinase inhibition attenuates oxidative stress in systemic immune cells and renal compartment during sepsis-induced acute kidney injury in mice. Int Immunopharmacol 2021; 90:107123. [PMID: 33168411 DOI: 10.1016/j.intimp.2020.107123] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022]
Abstract
Sepsis is a life-threatening condition which affects multiple organs including the kidney. Sepsis-induced acute kidney injury (AKI) is a major health burden throughout the globe. Pathogenesis of sepsis-induced AKI is complex; however, it involves both innate and adaptive immune cells such as B cells, T cells, dendritic cells (DCs), macrophages, and neutrophils. Bruton's tyrosine kinase (BTK) is reportedly involved in inflammatory and oxidative signaling in different immune cells, however its contribution with respect to sepsis-induced AKI has not been delineated. This study attempted to investigate the role of BTK and its inhibition on oxidizing enzymes NADPH oxidase (NOX-2) and inducible nitric oxide synthase (iNOS) in DCs, neutrophils, and B cells during AKI. Our data reveal that BTK is activated in DCs, neutrophils, and B cells which causes an increase in AKI associated biochemical markers such as serum creatinine/blood urea nitrogen, renal myeloperoxidase activity, and histopathological disturbances in renal tubular structures. Activation of BTK causes upregulation of NOX-2/iNOS/nitrotyrosine in these immune cells and kidney. Treatment with BTK inhibitor, Ibrutinib causes attenuation in AKI associated dysfunction in biochemical parameters (serum creatinine/blood urea nitrogen, renal myeloperoxidase activity) and oxidative stress in immune cells and kidney (iNOS/NOX2/lipid peroxides/nitrotyrosine/protein carbonyls). In summary, the current investigation reveals a compelling role of BTK signaling in sepsis-induced AKI which is evident from amelioration of AKI associated renal dysfunction after its inhibition.
Collapse
Affiliation(s)
- Ahmed Nadeem
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sheikh F Ahmad
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wael A Alanazi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hafiz M Mahmood
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sary Alsanea
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
179
|
Tan LL, Jiang XL, Xu LX, Li G, Feng CX, Ding X, Sun B, Qin ZH, Zhang ZB, Feng X, Li M. TP53-induced glycolysis and apoptosis regulator alleviates hypoxia/ischemia-induced microglial pyroptosis and ischemic brain damage. Neural Regen Res 2021; 16:1037-1043. [PMID: 33269748 PMCID: PMC8224121 DOI: 10.4103/1673-5374.300453] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Our previous studies have demonstrated that TP53-induced glycolysis and apoptosis regulator (TIGAR) can protect neurons after cerebral ischemia/reperfusion. However, the role of TIGAR in neonatal hypoxic-ischemic brain damage (HIBD) remains unknown. In the present study, 7-day-old Sprague-Dawley rat models of HIBD were established by permanent occlusion of the left common carotid artery followed by 2-hour hypoxia. At 6 days before induction of HIBD, a lentiviral vector containing short hairpin RNA of either TIGAR or gasdermin D (LV-sh_TIGAR or LV-sh_GSDMD) was injected into the left lateral ventricle and striatum. Highly aggressively proliferating immortalized (HAPI) microglial cell models of in vitro HIBD were established by 2-hour oxygen/glucose deprivation followed by 24-hour reoxygenation. Three days before in vitro HIBD induction, HAPI microglial cells were transfected with LV-sh_TIGAR or LV-sh_GSDMD. Our results showed that TIGAR expression was increased in the neonatal rat cortex after HIBD and in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Lentivirus-mediated TIGAR knockdown in rats markedly worsened pyroptosis and brain damage after hypoxia/ischemia in vivo and in vitro. Application of exogenous nicotinamide adenine dinucleotide phosphate (NADPH) increased the NADPH level and the glutathione/oxidized glutathione ratio and decreased reactive oxygen species levels in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Additionally, exogenous NADPH blocked the effects of TIGAR knockdown in neonatal HIBD in vivo and in vitro. These findings show that TIGAR can inhibit microglial pyroptosis and play a protective role in neonatal HIBD. The study was approved by the Animal Ethics Committee of Soochow University of China (approval No. 2017LW003) in 2017.
Collapse
Affiliation(s)
- Lan-Lan Tan
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiao-Lu Jiang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Li-Xiao Xu
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gen Li
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Chen-Xi Feng
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xin Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Bin Sun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zheng-Hong Qin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - Zu-Bin Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Mei Li
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
180
|
Mao L, Kitani A, Hiejima E, Montgomery-Recht K, Zhou W, Fuss I, Wiestner A, Strober W. Bruton tyrosine kinase deficiency augments NLRP3 inflammasome activation and causes IL-1β-mediated colitis. J Clin Invest 2020; 130:1793-1807. [PMID: 31895698 DOI: 10.1172/jci128322] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 12/23/2019] [Indexed: 12/22/2022] Open
Abstract
Bruton tyrosine kinase (BTK) is present in a wide variety of cells and may thus have important non-B cell functions. Here, we explored the function of this kinase in macrophages with studies of its regulation of the NLR family, pyrin domain-containing 3 (NLRP3) inflammasome. We found that bone marrow-derived macrophages (BMDMs) from BTK-deficient mice or monocytes from patients with X-linked agammaglobulinemia (XLA) exhibited increased NLRP3 inflammasome activity; this was also the case for BMDMs exposed to low doses of BTK inhibitors such as ibrutinib and for monocytes from patients with chronic lymphocytic leukemia being treated with ibrutinib. In mechanistic studies, we found that BTK bound to NLRP3 during the priming phase of inflammasome activation and, in doing so, inhibited LPS- and nigericin-induced assembly of the NLRP3 inflammasome during the activation phase of inflammasome activation. This inhibitory effect was caused by BTK inhibition of protein phosphatase 2A-mediated (PP2A-mediated) dephosphorylation of Ser5 in the pyrin domain of NLRP3. Finally, we show that BTK-deficient mice were subject to severe experimental colitis and that such colitis was normalized by administration of anti-IL-β or anakinra, an inhibitor of IL-1β signaling. Together, these studies strongly suggest that BTK functions as a physiologic inhibitor of NLRP3 inflammasome activation and explain why patients with XLA are prone to develop Crohn's disease.
Collapse
Affiliation(s)
- Liming Mao
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Atsushi Kitani
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Eitaro Hiejima
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Kim Montgomery-Recht
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research Inc., National Cancer Institute (NCI) Campus at Frederick, Frederick, Maryland, USA
| | - Wenchang Zhou
- Theoretical Molecular Biophysics Laboratory, National Heart, Lung and Blood Institute (NHLBI), and
| | - Ivan Fuss
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Adrian Wiestner
- Lymphoid Malignancies Section, Hematology Branch, NHLBI, NIH, Bethesda, Maryland, USA
| | - Warren Strober
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| |
Collapse
|
181
|
Feng H, Liu Y, Zhang R, Liang Y, Sun L, Lan N, Ma B. TSPO Ligands PK11195 and Midazolam Reduce NLRP3 Inflammasome Activation and Proinflammatory Cytokine Release in BV-2 Cells. Front Cell Neurosci 2020; 14:544431. [PMID: 33362467 PMCID: PMC7759202 DOI: 10.3389/fncel.2020.544431] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation related to microglial activation plays an important role in neurodegenerative diseases. Translocator protein 18 kDa (TSPO), a biomarker of reactive gliosis, its ligands can reduce neuroinflammation and can be used to treat neurodegenerative diseases. Therefore, we explored whether TSPO ligands exert an anti-inflammatory effect by affecting the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome, thereby inhibiting the release of inflammatory cytokines in microglial cells. In the present study, BV-2 cells were exposed to lipopolysaccharide (LPS) for 6 h to induce an inflammatory response. We found that the levels of reactive oxygen species (ROS), NLRP3 inflammasome, interleukin-1β (IL-1β), and interleukin-18 (IL-18) were significantly increased. However, pretreatment with TSPO ligands inhibited BV-2 microglial and NLRP3 inflammasome activation and significantly reduced the levels of ROS, IL-1β, and IL-18. Furthermore, a combination of LPS and ATP was used to activate the NLRP3 inflammasome. Both pretreatment and post-treatment with TSPO ligand can downregulate the activation of NLRP3 inflammasome and IL-1β expression. Finally, we found that TSPO was involved in the regulation of NLRP3 inflammasome with TSPO ligands treatment in TSPO knockdown BV2 cells. Collectively, these results indicate that TSPO ligands are promising targets to control microglial reactivity and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Hao Feng
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Yongxin Liu
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Rui Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Yingxia Liang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Lina Sun
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Nannan Lan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Baoyu Ma
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| |
Collapse
|
182
|
Schwaid AG, Spencer KB. Strategies for Targeting the NLRP3 Inflammasome in the Clinical and Preclinical Space. J Med Chem 2020; 64:101-122. [PMID: 33351619 DOI: 10.1021/acs.jmedchem.0c01307] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inhibiting the NLRP3 inflammasome mediates inflammation in an extensive number of preclinical models. As excitement in this field has grown, several companies have recently initiated testing of direct NLRP3 inhibitors in the clinic. At the same time, the NLRP3 inflammasome is part of a larger pro-inflammatory pathway, whose modulation is also being explored. Multiple targets in this pathway are already impinged upon by molecules that have been through clinical trials. These data, informed by the growing mechanistic understanding of the NLRP3 inflammasome in the preclinical space, provide a rich backdrop to assess the current state of the field. Here we explore attempts to inhibit the NLRP3 inflammasome in light of clinical and preclinical data around efficacy and safety.
Collapse
Affiliation(s)
- Adam G Schwaid
- Chemical Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Kerrie B Spencer
- Chemical Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
183
|
Zhang H, Zahid A, Ismail H, Tang Y, Jin T, Tao J. An overview of disease models for NLRP3 inflammasome over-activation. Expert Opin Drug Discov 2020; 16:429-446. [PMID: 33131335 DOI: 10.1080/17460441.2021.1844179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Inflammatory reactions, including those mediated by the NLRP3 inflammasome, maintain the body's homeostasis by removing pathogens, repairing damaged tissues, and adapting to stressed environments. However, uncontrolled activation of the NLRP3 inflammasome tends to cause various diseases using different mechanisms. Recently, many inhibitors of the NLRP3 inflammasome have been reported and many are being developed. In order to assess their efficacy, specificity, and mechanism of action, the screening process of inhibitors requires various types of cell and animal models of NLRP3-associated diseases.Areas covered: In the following review, the authors give an overview of the cell and animal models that have been used during the research and development of various inhibitors of the NLRP3 inflammasome.Expert opinion: There are many NLRP3 inflammasome inhibitors, but most of the inhibitors have poor specificity and often influence other inflammatory pathways. The potential risk for cross-reaction is high; therefore, the development of highly specific inhibitors is essential. The selection of appropriate cell and animal models, and combined use of different models for the evaluation of these inhibitors can help to clarify the target specificity and therapeutic effects, which is beneficial for the development and application of drugs targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ayesha Zahid
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hazrat Ismail
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science. Hefei National Science Center for Physical Sciences at Microscale. University of Science and Technology of China, Hefei, China
| | - Yujie Tang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Shanghai, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
184
|
Weber ANR, Bittner ZA, Shankar S, Liu X, Chang TH, Jin T, Tapia-Abellán A. Recent insights into the regulatory networks of NLRP3 inflammasome activation. J Cell Sci 2020; 133:133/23/jcs248344. [PMID: 33273068 DOI: 10.1242/jcs.248344] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is a fascinating cellular machinery endowed with the capacity for rapid proteolytic processing of the pro-inflammatory cytokine IL-1β and the cell death effector gasdermin D (GSDMD). Although its activity is essential to fight infection and support tissue homeostasis, the inflammasome complex, which consists of the danger sensor NLRP3, the adaptor apoptosis-associated speck-like protein containing a CARD (ASC; also known as PYCARD), caspase-1 and probably other regulatory proteins, also bears considerable potential for detrimental inflammation, as observed in human conditions such as gout, heart attack, stroke and Alzheimer's disease. Thus, multi-layered regulatory networks are required to ensure the fine balance between rapid responsiveness versus erroneous activation (sufficient and temporally restricted versus excessive and chronic activity) of the inflammasome. These involve multiple activation, secretion and cell death pathways, as well as modulation of the subcellular localization of NLRP3, and its structure and activity, owing to post-translational modification by other cellular proteins. Here, we discuss the exciting progress that has recently been made in deciphering the regulation of the NLRP3 inflammasome. Additionally, we highlight open questions and describe areas of research that warrant further exploration to obtain a more comprehensive molecular and cellular understanding of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Alexander N R Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany .,iFIT - Cluster of Excellence (EXC 2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University Hospital Tübingen - Internal Medicine VIII, Otfried-Müller-Str. 14, 72076 Tübingen, Germany
| | - Zsófia A Bittner
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Sangeetha Shankar
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Xiao Liu
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Tzu-Hsuan Chang
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Tengchuan Jin
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026 China
| | - Ana Tapia-Abellán
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| |
Collapse
|
185
|
Salina ACG, Brandt SL, Klopfenstein N, Blackman A, Bazzano JMR, Sá-Nunes A, Byers-Glosson N, Brodskyn C, Tavares NM, Da Silva IBS, Medeiros AI, Serezani CH. Leukotriene B 4 licenses inflammasome activation to enhance skin host defense. Proc Natl Acad Sci U S A 2020; 117:30619-30627. [PMID: 33184178 PMCID: PMC7720147 DOI: 10.1073/pnas.2002732117] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The initial production of inflammatory mediators dictates host defense as well as tissue injury. Inflammasome activation is a constituent of the inflammatory response by recognizing pathogen and host-derived products and eliciting the production of IL-1β and IL-18 in addition to inducing a type of inflammatory cell death termed "pyroptosis." Leukotriene B4 (LTB4) is a lipid mediator produced quickly (seconds to minutes) by phagocytes and induces chemotaxis, increases cytokine/chemokine production, and enhances antimicrobial effector functions. Whether LTB4 directly activates the inflammasome remains to be determined. Our data show that endogenously produced LTB4 is required for the expression of pro-IL-1β and enhances inflammasome assembly in vivo and in vitro. Furthermore, LTB4-mediated Bruton's tyrosine kinase (BTK) activation is required for inflammasome assembly in vivo as well for IL-1β-enhanced skin host defense. Together, these data unveil a new role for LTB4 in enhancing the expression and assembly of inflammasome components and suggest that while blocking LTB4 actions could be a promising therapeutic strategy to prevent inflammasome-mediated diseases, exogenous LTB4 can be used as an adjuvant to boost inflammasome-dependent host defense.
Collapse
Affiliation(s)
- Ana Carolina Guerta Salina
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Stephanie L Brandt
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-3082
| | - Nathan Klopfenstein
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Amondrea Blackman
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
| | | | - Anderson Sá-Nunes
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Nicole Byers-Glosson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-3082
| | - Claudia Brodskyn
- Oswaldo Cruz Foundation, Gonçalo Moniz Institute, FIOCRUZ, Salvador 40296-710, Brazil
| | | | | | - Alexandra I Medeiros
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil
| | - C Henrique Serezani
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232;
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
186
|
Chen T, Liu S, Zheng M, Li Y, He L. The effect of geniposide on chronic unpredictable mild stress‐induced depressive mice through
BTK
/
TLR4
/
NF‐κB
and
BDNF
/
TrkB
signaling pathways. Phytother Res 2020; 35:932-945. [DOI: 10.1002/ptr.6846] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Tong Chen
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Shengnan Liu
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Menglin Zheng
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Yixuan Li
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Ling He
- Department of Pharmacology China Pharmaceutical University Nanjing China
| |
Collapse
|
187
|
Varghese PM, Tsolaki AG, Yasmin H, Shastri A, Ferluga J, Vatish M, Madan T, Kishore U. Host-pathogen interaction in COVID-19: Pathogenesis, potential therapeutics and vaccination strategies. Immunobiology 2020; 225:152008. [PMID: 33130519 PMCID: PMC7434692 DOI: 10.1016/j.imbio.2020.152008] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023]
Abstract
The current coronavirus pandemic, COVID-19, is the third outbreak of disease caused by the coronavirus family, after Severe Acute Respiratory Syndrome and Middle East Respiratory Syndrome. It is an acute infectious disease caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This severe disease is characterised by acute respiratory distress syndrome, septic shock, metabolic acidosis, coagulation dysfunction, and multiple organ dysfunction syndromes. Currently, no drugs or vaccines exist against the disease and the only course of treatment is symptom management involving mechanical ventilation, immune suppressants, and repurposed drugs. The severe form of the disease has a relatively high mortality rate. The last six months have seen an explosion of information related to the host receptors, virus transmission, virus structure-function relationships, pathophysiology, co-morbidities, immune response, treatment and the most promising vaccines. This review takes a critically comprehensive look at various aspects of the host-pathogen interaction in COVID-19. We examine the genomic aspects of SARS-CoV-2, modulation of innate and adaptive immunity, complement-triggered microangiopathy, and host transmission modalities. We also examine its pathophysiological impact during pregnancy, in addition to emphasizing various gaps in our knowledge. The lessons learnt from various clinical trials involving repurposed drugs have been summarised. We also highlight the rationale and likely success of the most promising vaccine candidates.
Collapse
Affiliation(s)
- Praveen Mathews Varghese
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, London, United Kingdom; School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Anthony G Tsolaki
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, London, United Kingdom
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Abhishek Shastri
- Central and North West London NHS Foundation Trust, London, United Kingdom
| | - Janez Ferluga
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, London, United Kingdom
| | - Manu Vatish
- Department of Obstetrics and Gynaecology, Women's Centre, John Radcliffe Oxford University Hospital, Oxford, OX3 9DU, United Kingdom
| | - Taruna Madan
- Department of Innate Immunity, ICMR - National Institute for Research in Reproductive Health, J.M. Street, Parel, Mumbai, Maharashtra, India
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, London, United Kingdom.
| |
Collapse
|
188
|
Zhang C, Qin J, Zhang S, Zhang N, Tan B, Siwko S, Zhang Y, Wang Q, Chen J, Qian M, Liu M, Du B. ADP/P2Y 1 aggravates inflammatory bowel disease through ERK5-mediated NLRP3 inflammasome activation. Mucosal Immunol 2020; 13:931-945. [PMID: 32518369 DOI: 10.1038/s41385-020-0307-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 04/30/2020] [Accepted: 05/15/2020] [Indexed: 02/04/2023]
Abstract
Inflammasomes are essential for inflammation and pathogen elimination in response to microbial infection and endogenous danger signals. However, the mechanism of inflammasome activation by endogenous danger signals mediated posttranslational modification and the connection between inflammasomes and inflammatory diseases remains elusive. In this study, we found that ADP was highly released from injured colonic tissue as a danger signal during inflammatory bowel disease. Consequently, extracellular ADP activated the NLRP3 inflammasome through P2Y1 receptor-mediated calcium signaling, which led to the maturation and secretion of IL-1β and further aggravation of experimental colitis. Genetic ablation or pharmacological blockade of the P2Y1 receptor significantly ameliorated DSS-induced colitis and endotoxic shock through reducing NLRP3 inflammasome activation. Moreover, ERK5-mediated tyrosine phosphorylation of ASC was essential for activation of the NLRP3 inflammasome. Thus, our study provides a novel theoretical basis for posttranslational modification of ASC in NLRP3 inflammasome activation and revealed that ADP/P2Y1 is a potential drug target for inflammatory bowel disease.
Collapse
Affiliation(s)
- Chengfei Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China.,Department of Pathology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Juliang Qin
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China.,Joint Center for Translational Medicine, Fengxian District Central Hospital, No. 6600 Nanfeng Road, Fengxian District, Shanghai, 201499, China
| | - Su Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Na Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Binhe Tan
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Stefan Siwko
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, TX, 77030, USA
| | - Ying Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Qin Wang
- Joint Center for Translational Medicine, Fengxian District Central Hospital, No. 6600 Nanfeng Road, Fengxian District, Shanghai, 201499, China
| | - Jinlian Chen
- Joint Center for Translational Medicine, Fengxian District Central Hospital, No. 6600 Nanfeng Road, Fengxian District, Shanghai, 201499, China
| | - Min Qian
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Mingyao Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China.
| | - Bing Du
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
189
|
Bruton's Tyrosine Kinase Inhibitor Attenuates Warm Hepatic Ischemia/Reperfusion Injury via Modulation of the NLR Family Pyrin Domain Containing 3 Inflammasome. Transplant Proc 2020; 52:2947-2954. [PMID: 33131902 DOI: 10.1016/j.transproceed.2019.10.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/06/2019] [Indexed: 11/24/2022]
Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is a widely studied inflammasome that plays a critical role in inflammatory responses. Many triggers, including microbial pathogens (ie, bacteria and viruses) and other signals (ie, reactive oxygen species, adenosine triphosphate, urate, silicon, and asbestos), can stimulate the NLRP3 inflammasome. Liver ischemia/reperfusion (I/R) injury is a common pathologic process during liver surgery and shock and can induce severe liver damage. Although its pathogenesis is still unclear, oxidative stress and overproduction of the inflammatory response are likely to contribute to I/R injury. The NLRP3 inflammasome is activated during the I/R process, resulting in further recruitment and activation of caspase-1. Activated caspase-1 cleaves the pro-forms of interleukin-1β and interleukin-18 and results in their maturation, triggering a proinflammatory cytokine cascade and causing liver damage. Bruton's tyrosine kinase is a critical molecule involved in diverse cellular pathways, such as proliferation, apoptosis, inflammation, and angiogenesis. Intrahepatic Bruton's tyrosine kinase is mainly expressed on Kupffer cells and sinusoidal endothelial cells, and the inflammasome is activated in Kupffer cells. Our study found that inhibition of Bruton's tyrosine kinase effectively attenuated liver I/R injury by suppressing activation of the NLRP3 inflammasome in Kupffer cells.
Collapse
|
190
|
Wagatsuma K, Nakase H. Contradictory Effects of NLRP3 Inflammasome Regulatory Mechanisms in Colitis. Int J Mol Sci 2020; 21:ijms21218145. [PMID: 33143375 PMCID: PMC7662299 DOI: 10.3390/ijms21218145] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 02/08/2023] Open
Abstract
The inflammasome is an intracellular molecular complex, which is mainly involved in innate immunity. Inflammasomes are formed in response to danger signals, associated with infection and injury, and mainly regulate the secretion of interleukin-1β and interleukin-18. Inflammasome dysregulation is known to be associated with various diseases and conditions, and its regulatory mechanisms have become of great interest in recent years. In the colon, inflammasomes have been reported to be associated with autophagy and the microbiota, and their dysregulation contributes to colitis and. However, the detailed role of inflammasomes in inflammatory bowel disease is still under debate because the mechanisms that regulate the inflammasome are complex and the inflammasome components and cytokines show seemingly contradictory multiple effects. Herein, we comprehensively review the literature on inflammasome functioning in the colon and describe the complex interactions of the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome components with inflammatory cytokines, autophagy, and the microbiota in experimental colitis models and patients with inflammatory bowel disease.
Collapse
|
191
|
López-Reyes A, Martinez-Armenta C, Espinosa-Velázquez R, Vázquez-Cárdenas P, Cruz-Ramos M, Palacios-Gonzalez B, Gomez-Quiroz LE, Martínez-Nava GA. NLRP3 Inflammasome: The Stormy Link Between Obesity and COVID-19. Front Immunol 2020; 11:570251. [PMID: 33193349 PMCID: PMC7662564 DOI: 10.3389/fimmu.2020.570251] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Abstract
Several countries around the world have faced an important obesity challenge for the past four decades as the result of an obesogenic environment. This disease has a multifactorial origin and it is associated with multiple comorbidities including type 2 diabetes, hypertension, osteoarthritis, metabolic syndrome, cancer, and dyslipidemia. With regard to dyslipidemia, hypertriglyceridemia is a well-known activator of the NLRP3 inflammasome, triggering adipokines and cytokines secretion which in addition induce a systemic inflammatory state that provides an adequate scenario for infections, particularly those mediated by viruses such as HIV, H1N1 influenza, and SARS-CoV-2. The SARS-CoV-2 infection causes the coronavirus disease 2019 (COVID-19) and it is responsible for the pandemic that we are currently living. COVID-19 causes an aggressive immune response known as cytokine release syndrome or cytokine storm that causes multiorgan failure and in most cases leads to death. In the present work, we aimed to review the molecular mechanisms by which obesity-associated systemic inflammation could cause a more severe clinical presentation of COVID-19. The SARS-CoV-2 infection could potentiate or accelerate the pre-existing systemic inflammatory state of individuals with obesity, via the NLRP3 inflammasome activation and the release of pro-inflammatory cytokines from cells trough Gasdermin-pores commonly found in cell death by pyroptosis.
Collapse
Affiliation(s)
- Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, México
- Facultad de Ciencias de la Salud, Universidad Anáhuac, Ciudad de México, México
| | - Carlos Martinez-Armenta
- Postgrado en Biología Experimental, Dirección de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, México
| | | | - Paola Vázquez-Cárdenas
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, México
| | - Marlid Cruz-Ramos
- Cátedras de Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Cancerología, Ciudad de México, México
| | - Berenice Palacios-Gonzalez
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | - Luis Enrique Gomez-Quiroz
- Laboratorio de Fisiología Celular, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, México
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, México
| |
Collapse
|
192
|
O'Riordan CE, Purvis GSD, Collotta D, Krieg N, Wissuwa B, Sheikh MH, Ferreira Alves G, Mohammad S, Callender LA, Coldewey SM, Collino M, Greaves DR, Thiemermann C. X-Linked Immunodeficient Mice With No Functional Bruton's Tyrosine Kinase Are Protected From Sepsis-Induced Multiple Organ Failure. Front Immunol 2020; 11:581758. [PMID: 33162995 PMCID: PMC7580254 DOI: 10.3389/fimmu.2020.581758] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
We previously reported the Bruton's tyrosine kinase (BTK) inhibitors ibrutinib and acalabrutinib improve outcomes in a mouse model of polymicrobial sepsis. Now we show that genetic deficiency of the BTK gene alone in Xid mice confers protection against cardiac, renal, and liver injury in polymicrobial sepsis and reduces hyperimmune stimulation ("cytokine storm") induced by an overwhelming bacterial infection. Protection is due in part to enhanced bacterial phagocytosis in vivo, changes in lipid metabolism and decreased activation of NF-κB and the NLRP3 inflammasome. The inactivation of BTK leads to reduced innate immune cell recruitment and a phenotypic switch from M1 to M2 macrophages, aiding in the resolution of sepsis. We have also found that BTK expression in humans is increased in the blood of septic non-survivors, while lower expression is associated with survival from sepsis. Importantly no further reduction in organ damage, cytokine production, or changes in plasma metabolites is seen in Xid mice treated with the BTK inhibitor ibrutinib, demonstrating that the protective effects of BTK inhibitors in polymicrobial sepsis are mediated solely by inhibition of BTK and not by off-target effects of this class of drugs.
Collapse
Affiliation(s)
- Caroline E. O'Riordan
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Gareth S. D. Purvis
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Nadine Krieg
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Bianka Wissuwa
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Madeeha H. Sheikh
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | | - Shireen Mohammad
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Lauren A. Callender
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Christoph Thiemermann
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
193
|
Seoane PI, Lee B, Hoyle C, Yu S, Lopez-Castejon G, Lowe M, Brough D. The NLRP3-inflammasome as a sensor of organelle dysfunction. J Cell Biol 2020; 219:191204. [PMID: 33044555 PMCID: PMC7543090 DOI: 10.1083/jcb.202006194] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 02/08/2023] Open
Abstract
Diverse pathogen- and damage-associated stresses drive inflammation via activation of the multimolecular NLRP3-inflammasome complex. How the effects of diverse stimuli are integrated by the cell to regulate NLRP3 has been the subject of intense research, and yet an accepted unifying hypothesis for the control of NLRP3 remains elusive. Here, we review the literature on the effects of NLRP3-activating stimuli on subcellular organelles and conclude that a shared feature of NLRP3-activating stresses is an organelle dysfunction. In particular, we propose that the endosome may be more important than previously recognized as a signal-integrating hub for NLRP3 activation in response to many stimuli and may also link to the dysfunction of other organelles. In addition, NLRP3-inflammasome-activating stimuli trigger diverse posttranslational modifications of NLRP3 that are important in controlling its activation. Future research should focus on how organelles respond to specific NLRP3-activating stimuli, and how this relates to posttranslational modifications, to delineate the organellar control of NLRP3.
Collapse
Affiliation(s)
- Paula I. Seoane
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK,The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Bali Lee
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK,The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Christopher Hoyle
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK,The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Shi Yu
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK,The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Gloria Lopez-Castejon
- Division of Infection, Immunity, and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK,The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Martin Lowe
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - David Brough
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK,The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK,Correspondence to David Brough:
| |
Collapse
|
194
|
Martin E, Aigrot MS, Grenningloh R, Stankoff B, Lubetzki C, Boschert U, Zalc B. Bruton's Tyrosine Kinase Inhibition Promotes Myelin Repair. Brain Plast 2020; 5:123-133. [PMID: 33282676 PMCID: PMC7685672 DOI: 10.3233/bpl-200100] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Microglia are the resident macrophages of the central nervous system (CNS). In multiple sclerosis (MS) and related experimental models, microglia have either a pro-inflammatory or a pro-regenerative/pro-remyelinating function. Inhibition of Bruton’s tyrosine kinase (BTK), a member of the Tec family of kinases, has been shown to block differentiation of pro-inflammatory macrophages in response to granulocyte–macrophage colony-stimulating factor in vitro. However, the role of BTK in the CNS is unknown. Methods: Our aim was to investigate the effect of BTK inhibition on myelin repair in ex vivo and in vivo experimental models of demyelination and remyelination. The remyelination effect of a BTK inhibitor (BTKi; BTKi-1) was then investigated in LPC-induced demyelinated cerebellar organotypic slice cultures and metronidazole-induced demyelinated Xenopus MBP-GFP-NTR transgenic tadpoles. Results: Cellular detection of BTK and its activated form BTK-phospho-Y223 (p-BTK) was determined by immunohistochemistry in organotypic cerebellar slice cultures, before and after lysophosphatidylcholine (LPC)-induced demyelination. A low BTK signal detected by immunolabeling under normal conditions in cerebellar slices was in sharp contrast to an 8.5-fold increase in the number of BTK-positive cells observed in LPC-demyelinated slice cultures. Under both conditions, approximately 75% of cells expressing BTK and p-BTK were microglia and 25% were astrocytes. Compared with spontaneous recovery, treatment of demyelinated slice cultures and MTZ-demyelinated transgenic tadpoles with BTKi resulted in at least a 1.7-fold improvement of remyelination. Conclusion: Our data demonstrate that BTK inhibition is a promising therapeutic strategy for myelin repair.
Collapse
Affiliation(s)
- Elodie Martin
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière, GH Pitié-Salpêtrière, F-75013 Paris, France
| | - Marie-Stéphane Aigrot
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière, GH Pitié-Salpêtrière, F-75013 Paris, France
| | - Roland Grenningloh
- EMD Serono Research & Development Institute, Inc., Billerica, MA, United States (a business of Merck KGaA, Darmstadt, Germany)
| | - Bruno Stankoff
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière, GH Pitié-Salpêtrière, F-75013 Paris, France.,AP-HP, Saint-Antoine Hospital, F-75012 Paris, France
| | - Catherine Lubetzki
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière, GH Pitié-Salpêtrière, F-75013 Paris, France.,AP-HP, GH Pitié-Salpêtrière, F-75013 Paris, France
| | - Ursula Boschert
- Ares Trading S.A. an affiliate of Merck Serono S.A., Eysins, Switzerland
| | - Bernard Zalc
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière, GH Pitié-Salpêtrière, F-75013 Paris, France
| |
Collapse
|
195
|
Kong X, Liao Y, Zhou L, Zhang Y, Cheng J, Yuan Z, Wang S. Hematopoietic Cell Kinase (HCK) Is Essential for NLRP3 Inflammasome Activation and Lipopolysaccharide-Induced Inflammatory Response In Vivo. Front Pharmacol 2020; 11:581011. [PMID: 33041826 PMCID: PMC7523510 DOI: 10.3389/fphar.2020.581011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/20/2020] [Indexed: 01/15/2023] Open
Abstract
Activation of the NLRP3 inflammasome results in caspase 1 cleavage, which subsequently leads to IL-1β and IL-18 secretion, as well as pyroptosis, and aberrant activation of the inflammasome is involved in several diseases such as type 2 diabetes, atherosclerosis, multiple sclerosis, Parkinson's disease, and Alzheimer's disease. NLRP3 activity is regulated by various kinases. Genetic and pharmacological inhibition of the hematopoietic cell kinase (HCK), a member of the Src family of non-receptor tyrosine kinases (NRTKs) primarily expressed in myeloid cells, has previously been shown to ameliorate inflammation, indicating that it may be involved in the regulation of microglia function. However, the underlying mechanism is not known. Hence, in this study, we aimed to investigate the role of HCK in NLRP3 inflammasome activation. We demonstrated that HCK silencing inhibited NLRP3 inflammasome activation. Furthermore, the HCK-specific inhibitor, A419259, attenuated the release of IL-1β and caspase 1(P20) from the macrophages and microglia and reduced the formation of the apoptosis-associated speck-like protein with a CARD domain (ASC) oligomer. We also observed that HCK binds to full length NLRP3 and its NBD(NACHT) and LRR domains, but not to the PYD domain. In vivo, the HCK inhibitor attenuated the LPS-induced inflammatory response in the liver of LPS-challenged mice. Collectively, these results suggested that HCK plays a critical role in NLRP3 inflammasome activation. Our results will enhance current understanding regarding the effectiveness of HCK inhibitors for treating acute inflammatory diseases.
Collapse
Affiliation(s)
- Xiangxi Kong
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yajin Liao
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
| | - Lujun Zhou
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China.,Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Zhang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jinbo Cheng
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shukun Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
196
|
He J, Wu H, Zhou Y, Zheng C. Tomentosin inhibit cerebral ischemia/reperfusion induced inflammatory response via TLR4/ NLRP3 signalling pathway - in vivo and in vitro studies. Biomed Pharmacother 2020; 131:110697. [PMID: 32919189 DOI: 10.1016/j.biopha.2020.110697] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Stoke is a global threat, leading to 50 % of deaths worldwide and it causes permanent disability to about 5 million individuals globally each year. In this study, we assessed the potency of tomentosin to inhibit the neuroinflammation in in vivo and in vitro models. The Sprague Dawley rats were pretreated with 25 mg/kg bodyweight (b.wt) and 50 mg/kg b.wt of tomentosin for seven days followed by induction of cerebral ischemic reperfusion. The brain edema and cerebral infractions were analyzed. The levels of antioxidants and the interleukins were measured by standard methods. The NLRP3 signaling proteins expression was evaluated using qPCR analysis. In vitro studies were performed in SH-SY5Y-cells pretreated with tomentosin and subjected to OGD-R treatment. Our results depicts tomentosin scavenges the free radicals, enhances antioxidant system, inhibits the NLRP3 signaling. In vitro results substantiates with in vivo results. To conclude, our in vivo and in vitro results confirm tomentosin may be potent alternative for existing antistroke drugs.
Collapse
Affiliation(s)
- Jianguo He
- Department of Neurosurgery, Chongqing Red Cross Hospital (People's Hospital of Jiangbei District), Chongqing, 400020, China
| | - Haitao Wu
- Department of Neurosurgery, The First Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - YanYan Zhou
- Kuming Medical University Haiyuan College, Kunming, Yunnan, 651700, China
| | - Chao Zheng
- Department of Neurosurgery, Chongqing Red Cross Hospital (People's Hospital of Jiangbei District), Chongqing, 400020, China.
| |
Collapse
|
197
|
Okahara A, Koga JI, Matoba T, Fujiwara M, Tokutome M, Ikeda G, Nakano K, Tachibana M, Ago T, Kitazono T, Tsutsui H, Egashira K. Simultaneous targeting of mitochondria and monocytes enhances neuroprotection against ischemia-reperfusion injury. Sci Rep 2020; 10:14435. [PMID: 32879367 PMCID: PMC7468234 DOI: 10.1038/s41598-020-71326-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/10/2020] [Indexed: 01/15/2023] Open
Abstract
Ischemia-reperfusion injury impairs the efficacy of reperfusion therapy after ischemic stroke. Cyclophilin D (CypD)-mediated openings of mitochondrial permeability transition pore (mPTP) and subsequent monocyte-mediated inflammation are considered as major mechanisms of reperfusion injury. However, no medical therapies are currently available. Therefore, we have tested a hypothesis that simultaneous targeting of mPTP and inflammation confers substantial neuroprotection after cerebral ischemia-reperfusion. To address this point, we prepared CypD knockout mice, C-C chemokine receptor 2 (CCR2) knockout mice and CypD/CCR2 double knockout mice. These mice were subjected to 60 min transient cerebral ischemia by occluding middle cerebral arteries. Neurological deficits evaluated 3 days after reperfusion were significantly attenuated in CypD/CCR2 double knockout mice as compared to wild-type mice and other single knockout mice. Then, we have prepared polymeric nanoparticles containing cyclosporine A (CsA-NPs) and pitavastatin (Pitava-NPs), targeting mPTP opening and inflammation, respectively. Simultaneous administration of CsA-NP and Pitava-NP at the time of reperfusion also decreased infarct size and attenuated neurological deficits as compared to control nanoparticles and single administration of CsA-NPs or Pitava-NPs. These results indicate that simultaneous targeting of the mPTP opening and monocyte-mediated inflammation could be a novel strategy for better neurological outcomes in patients with ischemic stroke.
Collapse
Affiliation(s)
- Arihide Okahara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Jun-Ichiro Koga
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation, Kyushu University, Fukuoka, Japan.
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masaki Fujiwara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masaki Tokutome
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Gentaro Ikeda
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kaku Nakano
- Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation, Kyushu University, Fukuoka, Japan
| | - Masaki Tachibana
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kensuke Egashira
- Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation, Kyushu University, Fukuoka, Japan
- Department of Translational Medicine, Kyushu University Graduate School of Pharmaceutical Sciences, Fukuoka, Japan
| |
Collapse
|
198
|
Purvis GSD, Collino M, Aranda-Tavio H, Chiazza F, O'Riordan CE, Zeboudj L, Mohammad S, Collotta D, Verta R, Guisot NES, Bunyard P, Yaqoob MM, Greaves DR, Thiemermann C. Inhibition of Bruton's TK regulates macrophage NF-κB and NLRP3 inflammasome activation in metabolic inflammation. Br J Pharmacol 2020; 177:4416-4432. [PMID: 32608058 PMCID: PMC7484557 DOI: 10.1111/bph.15182] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/11/2020] [Accepted: 06/14/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE There are no medications currently available to treat metabolic inflammation. Bruton's tyrosine kinase (BTK) is highly expressed in monocytes and macrophages and regulates NF-κB and NLRP3 inflammasome activity; both propagate metabolic inflammation in diet-induced obesity. EXPERIMENTAL APPROACH Using an in vivo model of chronic inflammation, high-fat diet (HFD) feeding, in male C57BL/6J mice and in vitro assays in primary murine and human macrophages, we investigated if ibrutinib, an FDA approved BTK inhibitor, may represent a novel anti-inflammatory medication to treat metabolic inflammation. KEY RESULTS HFD-feeding was associated with increased BTK expression and activation, which was significantly correlated with monocyte/macrophage accumulation in the liver, adipose tissue, and kidney. Ibrutinib treatment to HFD-fed mice inhibited the activation of BTK and reduced monocyte/macrophage recruitment to the liver, adipose tissue, and kidney. Ibrutinib treatment to HFD-fed mice decreased the activation of NF-κB and the NLRP3 inflammasome. As a result, ibrutinib treated mice fed HFD had improved glycaemic control through restored signalling by the IRS-1/Akt/GSK-3β pathway, protecting mice against the development of hepatosteatosis and proteinuria. We show that BTK regulates NF-κB and the NLRP3 inflammasome specifically in primary murine and human macrophages, the in vivo cellular target of ibrutinib. CONCLUSION AND IMPLICATIONS We provide "proof of concept" evidence that BTK is a novel therapeutic target for the treatment of diet-induced metabolic inflammation and ibrutinib may be a candidate for drug repurposing as an anti-inflammatory agent for the treatment of metabolic inflammation in T2D and microvascular disease.
Collapse
Affiliation(s)
- Gareth S D Purvis
- William Harvey Research Institute, Queen Mary University of London, London, UK.,Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | | | - Fausto Chiazza
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | | | - Lynda Zeboudj
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Shireen Mohammad
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Roberta Verta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | | | | | - Magdi M Yaqoob
- William Harvey Research Institute, Queen Mary University of London, London, UK.,Centre for Diabetic Kidney Disease, Bart's and The London Hospital, London, UK
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Christoph Thiemermann
- William Harvey Research Institute, Queen Mary University of London, London, UK.,Centre for Diabetic Kidney Disease, Bart's and The London Hospital, London, UK
| |
Collapse
|
199
|
Negishi Y, Shima Y, Takeshita T, Morita R. Harmful and beneficial effects of inflammatory response on reproduction: sterile and pathogen-associated inflammation. Immunol Med 2020; 44:98-115. [PMID: 32838688 DOI: 10.1080/25785826.2020.1809951] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In reproduction, inflammatory processes play important roles in the development of many pregnancy complications such as preterm labor/birth, recurrent pregnancy loss, recurrent implantation failure, and preeclampsia. Inflammation can be initiated by both microbial and non-microbial causes. Bacterial infection in the feto-maternal interface and uterus can provoke preterm labor/birth, miscarriage, and chronic endometritis. By contrast, inflammation without infection, or 'sterile inflammation,' can also lead to many kinds of complications, such as preterm labor/birth, miscarriage, or preeclampsia. Aberrant inflammation is facilitated by immune cells such as macrophages, dendritic cells, natural killer cells, and invariant natural killer T cells. In addition, cytokines, chemokines, and several kinds of inflammatory mediators are involved. On the other hand, appropriate inflammation is required for a successful offspring during the progression of the entire pregnancy. Herein, we discuss the relation between pregnancy and inflammation with immunological alterations. Understanding the role of inflammation in complications during pregnancy may establish new perspectives of the progress of normal pregnancy as well as treatments during pregnancy complications.
Collapse
Affiliation(s)
- Yasuyuki Negishi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan.,Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo, Japan
| | - Yoshio Shima
- Department of Pediatrics, Nippon Medical School Musashikosugi Hospital, Kanagawa, Japan
| | - Toshiyuki Takeshita
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
200
|
IL-37 Gene Modification Enhances the Protective Effects of Mesenchymal Stromal Cells on Intestinal Ischemia Reperfusion Injury. Stem Cells Int 2020; 2020:8883636. [PMID: 32849879 PMCID: PMC7439787 DOI: 10.1155/2020/8883636] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/15/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Background Ischemia reperfusion injury (IRI) is the major cause of intestinal damage in clinic. Although either mesenchymal stromal cells (MSCs) or interleukin 37 (IL-37) shows some beneficial roles to ameliorate IRI, their effects are limited. In this study, the preventative effects of IL-37 gene-modified MSCs (IL-37-MSCs) on intestinal IRI are investigated. Methods Intestinal IRI model was established by occluding the superior mesenteric artery for 30 minutes and then reperfused for 72 hours in rats. Forty adult male Sprague-Dawley rats were randomly divided into the sham control, IL-37-MSC-treated, MSC-treated, recombinant IL-37- (rIL-37-) treated, and untreated groups. Intestinal damage was assessed by H&E staining. The levels of gut barrier function factors (diamine oxidase and D-Lactate) and inflammation cytokine IL-1β were assayed using ELISA. The synthesis of tissue damage-related NLRP3 inflammasome and downstream cascade reactions including cleaved caspase-1, IL-1β, and IL-18 was detected by western blot. The mRNA levels of proinflammatory mediators IL-6 and TNF-α, which are downstream of IL-1β and IL-18, were determined by qPCR. Data were analyzed by one-way analysis of variance (ANOVA) after the normality test and followed by post hoc analysis with the least significant difference (LSD) test. Results IL-37-MSCs were able to migrate to the damaged tissue and significantly inhibit intestinal IRI. As compared with MSCs or the rIL-37 monotherapy group, IL-37-MSC treatment both improved gut barrier function and decreased local and systemic inflammation cytokine IL-1β level in IRI rats. In addition, tissue damage-related NLRP3 and downstream targets (cleaved caspase-1, IL-1β, and IL-18) were significantly decreased in IRI rats treated with IL-37-MSCs. Furthermore, IL-1β- and IL-18-related proinflammatory mediator IL-6 and TNF-α mRNA expressions were all significantly decreased in IRI rats treated with IL-37-MSCs. Conclusion The results suggest that IL-37 gene modification significantly enhances the protective effects of MSCs against intestinal IRI. In addition, NLRP3-related signaling pathways could be associated with IL-37-MSC-mediated protection.
Collapse
|