1
|
Bruins WSC, Rentenaar R, Newcomb J, Zheng W, Ruiter RWJ, Baardemans T, Poma E, Moore C, Robinson GL, Lublinsky A, Zhang Y, Syed S, Milhollen M, Dash AB, van de Donk NWCJ, Groen RWJ, Zweegman S, Mutis T. Preclinical evaluation of the CD38-targeting engineered toxin body MT-0169 against multiple myeloma. Hemasphere 2024; 8:e70039. [PMID: 39544624 PMCID: PMC11561653 DOI: 10.1002/hem3.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/04/2024] [Accepted: 09/28/2024] [Indexed: 11/17/2024] Open
Abstract
Despite significant progress in the treatment of multiple myeloma (MM), relapsed/refractory patients urgently require more effective therapies. We here describe the discovery, mechanism of action, and preclinical anti-MM activity of engineered toxin body MT-0169, a next-generation immunotoxin comprising a CD38-specific antibody fragment linked to a de-immunized Shiga-like toxin A subunit (SLTA) payload. We show that specific binding of MT-0169 to CD38 on MM cell lines triggers rapid internalization of SLTA, causing cell death via irreversible ribosome inhibition, protein synthesis blockade, and caspase 3/7 activation. In co-culture experiments, bone marrow mesenchymal stromal cells did not induce drug resistance against MT-0169. In the preclinical setting, MT-0169 effectively lysed primary MM cells from newly diagnosed and heavily pretreated MM patients, including those refractory to daratumumab, with minimal toxicity against nonmalignant hematopoietic cells. MM cell lysis showed a significant correlation with their CD38 expression levels but not with cytogenetic risk, tumor load, or number of prior lines of therapy. Finally, MT-0169 showed efficient in vivo anti-MM activity in various mouse xenograft models, including one in which MM cells are grown in a humanized bone marrow-like niche. These findings support clinical investigation of MT-0169 in relapsed/refractory MM patients, including those refractory to CD38-targeting immunotherapies.
Collapse
Affiliation(s)
- Wassilis S. C. Bruins
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Rosa Rentenaar
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - John Newcomb
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Wenrou Zheng
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Ruud W. J. Ruiter
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Thomas Baardemans
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Eric Poma
- Molecular Templates, Inc.AustinTexasUSA
| | | | | | - Anya Lublinsky
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Yuhong Zhang
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Sakeena Syed
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | | | - Ajeeta B. Dash
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Niels W. C. J. van de Donk
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Richard W. J. Groen
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Sonja Zweegman
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| | - Tuna Mutis
- Amsterdam UMC location Vrije Universiteit Amsterdam, HematologyAmsterdamNetherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, HematologyAmsterdamNetherlands
| |
Collapse
|
2
|
Xiang X, Huang Y, Shen Y, Lv J, Li W, Dong M, Sun Y, Xu J, Cui M, Huang Y, Xia J. Radix Isatidis polysaccharide (RIP) alleviates QX-genotype infectious bronchitis virus-induced interstitial nephritis through the Nrf2/NLRP3/Caspase-3 signaling pathway. Int J Biol Macromol 2024; 278:134571. [PMID: 39147344 DOI: 10.1016/j.ijbiomac.2024.134571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/05/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Interstitial nephritis is the primary cause of mortality in IBV-infected chickens. Our previous research has demonstrated that Radix Isatidis polysaccharide (RIP) could alleviate this form of interstitial nephritis. To explore the mechanism, SPF chickens and chicken embryonic kidney cells (CEKs) were pre-treated with RIP and subsequently infected with QX-genotype IBV strain. Kidneys were sampled for transcriptomic and metabolomic analyses, and the cecum contents were collected for 16S rRNA gene sequencing. Results showed that pre-treatment with RIP led to a 50 % morbidity reduction in infected-chickens, along with decreased tissue lesion and viral load in the kidneys. Multi-omics analysis indicated three possible pathways (including antioxidant, anti-inflammatory and anti-apoptosis) which associated with RIP's efficacy against interstitial nephritis. Following further validation both in vivo and in vitro, the results showed that pre-treatment with RIP could activate the antioxidant transcription factor Nrf2, stimulate antioxidant enzyme expression, and consequently inhibit oxidative stress. Pre-treatment with RIP could also significantly reduce the expression of NLRP3 inflammasome and apoptosis-associated proteins (including Bax, Caspase-3, and Caspase-9). Additionally, RIP was also observed to promote the growth of beneficial bacteria in the intestine. Overall, pretreatment with RIP can alleviate QX-genotype IBV-induced interstitial nephritis via the Nrf2/NLRP3/Caspase-3 signaling pathway. This study lays the groundwork for the potential use of RIP in controlling avian infectious bronchitis (IB).
Collapse
Affiliation(s)
- Xuelian Xiang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Yamei Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Yuxi Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Jiadai Lv
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Wenwen Li
- Agricultural Service Center, Shanghe Town, Tongnan District, Chongqing 402671, China
| | - Mengyi Dong
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Yi Sun
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Jing Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Min Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China
| | - Jing Xia
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China; Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan 611130, China.
| |
Collapse
|
3
|
Pan Y, Li J, Lin P, Wan L, Qu Y, Cao L, Wang L. A review of the mechanisms of abnormal ceramide metabolism in type 2 diabetes mellitus, Alzheimer's disease, and their co-morbidities. Front Pharmacol 2024; 15:1348410. [PMID: 38379904 PMCID: PMC10877008 DOI: 10.3389/fphar.2024.1348410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
The global prevalence of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) is rapidly increasing, revealing a strong association between these two diseases. Currently, there are no curative medication available for the comorbidity of T2DM and AD. Ceramides are structural components of cell membrane lipids and act as signal molecules regulating cell homeostasis. Their synthesis and degradation play crucial roles in maintaining metabolic balance in vivo, serving as important mediators in the development of neurodegenerative and metabolic disorders. Abnormal ceramide metabolism disrupts intracellular signaling, induces oxidative stress, activates inflammatory factors, and impacts glucose and lipid homeostasis in metabolism-related tissues like the liver, skeletal muscle, and adipose tissue, driving the occurrence and progression of T2DM. The connection between changes in ceramide levels in the brain, amyloid β accumulation, and tau hyper-phosphorylation is evident. Additionally, ceramide regulates cell survival and apoptosis through related signaling pathways, actively participating in the occurrence and progression of AD. Regulatory enzymes, their metabolites, and signaling pathways impact core pathological molecular mechanisms shared by T2DM and AD, such as insulin resistance and inflammatory response. Consequently, regulating ceramide metabolism may become a potential therapeutic target and intervention for the comorbidity of T2DM and AD. The paper comprehensively summarizes and discusses the role of ceramide and its metabolites in the pathogenesis of T2DM and AD, as well as the latest progress in the treatment of T2DM with AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lei Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Singh S, Sharma S, Sharma H. Potential Impact of Bioactive Compounds as NLRP3 Inflammasome Inhibitors: An Update. Curr Pharm Biotechnol 2024; 25:1719-1746. [PMID: 38173061 DOI: 10.2174/0113892010276859231125165251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 01/05/2024]
Abstract
The inflammasome NLRP3 comprises a caspase recruitment domain, a pyrin domain containing receptor 3, an apoptosis-linked protein like a speck containing a procaspase-1, and an attached nucleotide domain leucine abundant repeat. There are a wide variety of stimuli that can activate the inflammasome NLRP3. When activated, the protein NLRP3 appoints the adapter protein ASC. Adapter ASC protein then recruits the procaspase-1 protein, which causes the procaspase- 1 protein to be cleaved and activated, which induces cytokines. At the same time, abnormal activation of inflammasome NLRP3 is associated with many diseases, such as diabetes, atherosclerosis, metabolic syndrome, cardiovascular and neurodegenerative diseases. As a result, a significant amount of effort has been put into comprehending the mechanisms behind its activation and looking for their specific inhibitors. In this review, we primarily focused on phytochemicals that inhibit the inflammasome NLRP3, as well as discuss the defects caused by NLRP3 signaling. We conducted an in-depth research review by searching for relevant articles in the Scopus, Google Scholar, and PubMed databases. By gathering information on phytochemical inhibitors that block NLRP3 inflammasome activation, a complicated balance between inflammasome activation or inhibition with NLRP3 as a key role was revealed in NLRP3-driven clinical situations.
Collapse
Affiliation(s)
- Sonia Singh
- Department of Pharmacy, Institute of Pharmaceutical Research, GLA University, Uttar Pradesh-281406, India
| | - Shiwangi Sharma
- Department of Pharmacy, Institute of Pharmaceutical Research, GLA University, Uttar Pradesh-281406, India
| | - Himanshu Sharma
- Department of Computer Engineering & Applications, GLA University, Uttar Pradesh-281406, India
| |
Collapse
|
5
|
Peng T, Zhang C, Chen WJ, Zhao XF, Wu WB, Yang WJ, Liang RJ. Pyroptosis: the dawn of a new era in endometrial cancer treatment. Front Oncol 2023; 13:1277639. [PMID: 37965452 PMCID: PMC10642841 DOI: 10.3389/fonc.2023.1277639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Endometrial cancer (EC) is a malignancy of the inner epithelial lining of the uterus. While early-stage EC is often curable through surgery, the management of advanced, recurrent and metastatic EC poses significant challenges and is associated with a poor prognosis. Pyroptosis, an emerging form of programmed cell death, is characterized by the cleavage of gasdermin proteins, inducing the formation of extensive gasdermin pores in the cell membrane and the leakage of interleukin-1β (IL-1β) and interleukin-18 (IL-18), consequently causing cell swelling, lysis and death. It has been found to be implicated in the occurrence and progression of almost all tumors. Recent studies have demonstrated that regulating tumor cells pyroptosis can exploit synergies function with traditional tumor treatments. This paper provides an overview of the research progress made in molecular mechanisms of pyroptosis. It then discusses the role of pyroptosis and its components in initiation and progression of endometrial cancer, emphasizing recent insights into the underlying mechanisms and highlighting unresolved questions. Furthermore, it explores the potential value of pyroptosis in the treatment of endometrial cancer, considering its current application in tumor radiotherapy, chemotherapy, targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Tian Peng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Zhang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wen-Jun Chen
- School of Nursing, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Gynaecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xue-Fei Zhao
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei-Bo Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei-Ji Yang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruo-Jia Liang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gynaecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Tang J, Lu X, Zhang T, Feng Y, Xu Q, Li J, Lan Y, Luo H, Zeng L, Xiang Y, Zhang Y, Li Q, Mao X, Tang B, Zeng D. Shiga toxin 2 A-subunit induces mitochondrial damage, mitophagy and apoptosis via the interaction of Tom20 in Caco-2 cells. Heliyon 2023; 9:e20012. [PMID: 37809632 PMCID: PMC10559750 DOI: 10.1016/j.heliyon.2023.e20012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/18/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Shiga toxin type 2 (Stx2) is the primary virulence factor produced by Shiga toxin-producing enterohemorrhagic Escherichia coli (STEC), which causes epidemic outbreaks of gastrointestinal sickness and potentially fatal sequela hemolytic uremic syndrome (HUS). Most studies on Stx2-induced apoptosis have been performed with holotoxins, but the mechanism of how the A and B subunits of Stx2 cause apoptosis in cells is not clear. Here, we found that Stx2 A-subunit (Stx2A) induced mitochondrial damage, PINK1/Parkin-dependent mitophagy and apoptosis in Caco-2 cells. PINK1/Parkin-dependent mitophagy caused by Stx2A reduced apoptosis by decreasing the accumulation of reactive oxidative species (ROS). Mechanistically, Stx2A interacts with Tom20 on mitochondria to initiate the translocation of Bax to mitochondria, leading to mitochondrial damage and apoptosis. Overall, these data suggested that Stx2A induces mitochondrial damage, mitophagy and apoptosis via the interaction of Tom20 in Caco-2 cells and that mitophagy caused by Stx2A ameliorates apoptosis by eliminating damaged mitochondria. These findings provide evidence for the potential use of Tom20 inhibition as an anti-Shiga toxin therapy.
Collapse
Affiliation(s)
- Jie Tang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Xiaoxue Lu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Tao Zhang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yuyang Feng
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qiaolin Xu
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Jing Li
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yuanzhi Lan
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Huaxing Luo
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Linghai Zeng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yuanyuan Xiang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yan Zhang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xuhu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Bin Tang
- Department of Clinical Laboratory, Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin, Chongqing, 402260, China
| | - Dongzhu Zeng
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| |
Collapse
|
7
|
Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol 2023; 16:24. [PMID: 36932407 PMCID: PMC10022228 DOI: 10.1186/s13045-023-01407-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/08/2023] [Indexed: 03/19/2023] Open
Abstract
Inflammasomes are macromolecular platforms formed in response to damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns, whose formation would cause maturation of interleukin-1 (IL-1) family members and gasdermin D (GSDMD), leading to IL-1 secretion and pyroptosis respectively. Several kinds of inflammasomes detecting different types of dangers have been found. The activation of inflammasomes is regulated at both transcription and posttranscription levels, which is crucial in protecting the host from infections and sterile insults. Present findings have illustrated that inflammasomes are involved in not only infection but also the pathology of tumors implying an important link between inflammation and tumor development. Generally, inflammasomes participate in tumorigenesis, cell death, metastasis, immune evasion, chemotherapy, target therapy, and radiotherapy. Inflammasome components are upregulated in some tumors, and inflammasomes can be activated in cancer cells and other stromal cells by DAMPs, chemotherapy agents, and radiation. In some cases, inflammasomes inhibit tumor progression by initiating GSDMD-mediated pyroptosis in cancer cells and stimulating IL-1 signal-mediated anti-tumor immunity. However, IL-1 signal recruits immunosuppressive cell subsets in other cases. We discuss the conflicting results and propose some possible explanations. Additionally, we also summarize interventions targeting inflammasome pathways in both preclinical and clinical stages. Interventions targeting inflammasomes are promising for immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Ziqi Zhang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xue Li
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| |
Collapse
|
8
|
Li MC, Tian Q, Liu S, Han SM, Zhang W, Qin XY, Chen JH, Liu CL, Guo YJ. The mechanism and relevant mediators associated with neuronal apoptosis and potential therapeutic targets in subarachnoid hemorrhage. Neural Regen Res 2023; 18:244-252. [PMID: 35900398 PMCID: PMC9396483 DOI: 10.4103/1673-5374.346542] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a dominant cause of death and disability worldwide. A sharp increase in intracranial pressure after SAH leads to a reduction in cerebral perfusion and insufficient blood supply for neurons, which subsequently promotes a series of pathophysiological responses leading to neuronal death. Many previous experimental studies have reported that excitotoxicity, mitochondrial death pathways, the release of free radicals, protein misfolding, apoptosis, necrosis, autophagy, and inflammation are involved solely or in combination in this disorder. Among them, irreversible neuronal apoptosis plays a key role in both short- and long-term prognoses after SAH. Neuronal apoptosis occurs through multiple pathways including extrinsic, mitochondrial, endoplasmic reticulum, p53 and oxidative stress. Meanwhile, a large number of blood contents enter the subarachnoid space after SAH, and the secondary metabolites, including oxygenated hemoglobin and heme, further aggravate the destruction of the blood-brain barrier and vasogenic and cytotoxic brain edema, causing early brain injury and delayed cerebral ischemia, and ultimately increasing neuronal apoptosis. Even there is no clear and effective therapeutic strategy for SAH thus far, but by understanding apoptosis, we might excavate new ideas and approaches, as targeting the upstream and downstream molecules of apoptosis-related pathways shows promise in the treatment of SAH. In this review, we summarize the existing evidence on molecules and related drugs or molecules involved in the apoptotic pathway after SAH, which provides a possible target or new strategy for the treatment of SAH.
Collapse
|
9
|
Yin H, Liu N, Sigdel KR, Duan L. Role of NLRP3 Inflammasome in Rheumatoid Arthritis. Front Immunol 2022; 13:931690. [PMID: 35833125 PMCID: PMC9271572 DOI: 10.3389/fimmu.2022.931690] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by multi-articular, symmetrical and invasive arthritis resulting from immune system abnormalities involving T and B lymphocytes. Although significant progress has been made in the understanding of RA pathogenesis, the underlying mechanisms are not fully understood. Recent studies suggest that NLRP3 inflammasome, a regulator of inflammation, might play an important role in the development of RA. There have been increasing clinical and pre-clinical evidence showing the treatment of NLRP3/IL-1β in inflammatory diseases. To provide a foundation for the development of therapeutic strategies, we will briefly summarize the roles of NLRP3 inflammasome in RA and explore its potential clinical treatment.
Collapse
Affiliation(s)
- Hui Yin
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Na Liu
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Keshav Raj Sigdel
- Department of Internal Medicine, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Lihua Duan
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- *Correspondence: Lihua Duan,
| |
Collapse
|
10
|
Gencturk E, Kasim M, Morova B, Kiraz A, Ulgen KO. Understanding the Link between Inflammasome and Apoptosis through the Response of THP-1 Cells against Drugs Using Droplet-Based Microfluidics. ACS OMEGA 2022; 7:16323-16332. [PMID: 35601322 PMCID: PMC9118214 DOI: 10.1021/acsomega.1c06569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/20/2022] [Indexed: 05/09/2023]
Abstract
Droplet-based microfluidic devices are used to investigate monocytic THP-1 cells in response to drug administration. Consistent and reproducible droplets are created, each of which acts as a bioreactor to carry out single cell experiments with minimized contamination and live cell tracking under an inverted fluorescence microscope for more than 2 days. Here, the effects of three different drugs (temsirolimus, rifabutin, and BAY 11-7082) on THP-1 are examined and the results are analyzed in the context of the inflammasome and apoptosis relationship. The ASC adaptor gene tagged with GFP is monitored as the inflammasome reporter. Thus, a systematic way is presented for deciphering cell-to-cell heterogeneity, which is an important issue in cancer treatment. The drug temsirolimus, which has effects of disrupting the mTOR pathway and triggering apoptosis in tumor cells, causes THP-1 cells to express ASC and to be involved in apoptosis. Treatment with rifabutin, which inhibits proliferation and initiates apoptosis in cells, affects ASC expression by first increasing and then decreasing it. CASP-3, which has a role in apoptosis and is directly related to ASC, has an increasing level in inflammasome conditioning. Thus, the cell under the effect of rifabutin might be faced with programmed cell death faster. The drug BAY 11-7082, which is responsible for NFκB inhibition, shows similar results to temsirolimus with more than 60% of cells having high fluorescence intensity (ASC expression). The microfluidic platform presented here offers strong potential for studying newly developed small-molecule inhibitors for personalized/precision medicine.
Collapse
Affiliation(s)
- Elif Gencturk
- Department
of Chemical Engineering, Boǧaziçi
University, Biosystems Engineering Laboratory, Istanbul 34342, Turkey
| | - Muge Kasim
- Department
of Chemical Engineering, Boǧaziçi
University, Biosystems Engineering Laboratory, Istanbul 34342, Turkey
| | - Berna Morova
- Department
of Physics, Koç University, Sariyer, 34450 Istanbul, Turkey
| | - Alper Kiraz
- Department
of Physics, Koç University, Sariyer, 34450 Istanbul, Turkey
- Department
of Electrical and Electronics Engineering, Koç University, Sariyer, 34450 Istanbul, Turkey
| | - Kutlu O. Ulgen
- Department
of Chemical Engineering, Boǧaziçi
University, Biosystems Engineering Laboratory, Istanbul 34342, Turkey
| |
Collapse
|
11
|
Zhang Y, Xing CJ, Liu X, Li YH, Jia J, Feng JG, Yang CJ, Chen Y, Zhou J. Thioredoxin-Interacting Protein (TXNIP) Knockdown Protects against Sepsis-Induced Brain Injury and Cognitive Decline in Mice by Suppressing Oxidative Stress and Neuroinflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8645714. [PMID: 35571246 PMCID: PMC9098358 DOI: 10.1155/2022/8645714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/17/2022] [Accepted: 04/23/2022] [Indexed: 11/19/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is linked to increased morbidity and mortality rates in patients with sepsis. Increased cytokine production and neuronal apoptosis are implicated in the pathogenesis of the SAE. Neuroinflammation plays a major role in sepsis-induced brain injury. Thioredoxin-interacting protein (TXNIP), an inhibitor of thioredoxin, is associated with oxidative stress and inflammation. However, whether the TXNIP is involved in the sepsis-induced brain injury and the underlying mechanism is yet to be elucidated. Therefore, the present study was aimed at elucidating the effects of TXNIP knockdown on sepsis-induced brain injury and cognitive decline in mice. Lipopolysaccharide (LPS) was injected intraperitoneally to induce sepsis brain injury in mice. The virus-carrying control or TXNIP shRNA was injected into the lateral ventricle of the brain 4 weeks before the LPS treatment. The histological changes in the hippocampal tissues, encephaledema, and cognitive function were detected, respectively. Also, the 7-day survival rate was recorded. Furthermore, the alterations in microglial activity, oxidative response, proinflammatory factors, apoptosis, protein levels (TXNIP and NLRP3 inflammasome), and apoptosis were examined in the hippocampal tissues. The results demonstrated that the TXNIP and NLRP3 inflammasome expression levels were increased at 6, 12, and 24 h post-LPS injection. TXNIP knockdown dramatically ameliorated the 7-day survival rate, cognitive decline, brain damage, neuronal apoptosis, and the brain water content, inhibited the activation of microglia, downregulated the NLRP3/caspase-1 signaling pathway, and reduced the oxidative stress and the neuroinflammatory cytokine levels at 24 h post-LPS injection. These results suggested a crucial effect of TXNIP knockdown on the mechanism of brain injury and cognitive decline in sepsis mice via suppressing oxidative stress and neuroinflammation. Thus, TXNIP might be a potential therapeutic target for SAE patients.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Cheng-Jun Xing
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiao Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ya-Hong Li
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Jian-Guo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Cheng-Jie Yang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Alijagic A, Engwall M, Särndahl E, Karlsson H, Hedbrant A, Andersson L, Karlsson P, Dalemo M, Scherbak N, Färnlund K, Larsson M, Persson A. Particle Safety Assessment in Additive Manufacturing: From Exposure Risks to Advanced Toxicology Testing. FRONTIERS IN TOXICOLOGY 2022; 4:836447. [PMID: 35548681 PMCID: PMC9081788 DOI: 10.3389/ftox.2022.836447] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Additive manufacturing (AM) or industrial three-dimensional (3D) printing drives a new spectrum of design and production possibilities; pushing the boundaries both in the application by production of sophisticated products as well as the development of next-generation materials. AM technologies apply a diversity of feedstocks, including plastic, metallic, and ceramic particle powders with distinct size, shape, and surface chemistry. In addition, powders are often reused, which may change the particles' physicochemical properties and by that alter their toxic potential. The AM production technology commonly relies on a laser or electron beam to selectively melt or sinter particle powders. Large energy input on feedstock powders generates several byproducts, including varying amounts of virgin microparticles, nanoparticles, spatter, and volatile chemicals that are emitted in the working environment; throughout the production and processing phases. The micro and nanoscale size may enable particles to interact with and to cross biological barriers, which could, in turn, give rise to unexpected adverse outcomes, including inflammation, oxidative stress, activation of signaling pathways, genotoxicity, and carcinogenicity. Another important aspect of AM-associated risks is emission/leakage of mono- and oligomers due to polymer breakdown and high temperature transformation of chemicals from polymeric particles, both during production, use, and in vivo, including in target cells. These chemicals are potential inducers of direct toxicity, genotoxicity, and endocrine disruption. Nevertheless, understanding whether AM particle powders and their byproducts may exert adverse effects in humans is largely lacking and urges comprehensive safety assessment across the entire AM lifecycle-spanning from virgin and reused to airborne particles. Therefore, this review will detail: 1) brief overview of the AM feedstock powders, impact of reuse on particle physicochemical properties, main exposure pathways and protective measures in AM industry, 2) role of particle biological identity and key toxicological endpoints in the particle safety assessment, and 3) next-generation toxicology approaches in nanosafety for safety assessment in AM. Altogether, the proposed testing approach will enable a deeper understanding of existing and emerging particle and chemical safety challenges and provide a strategy for the development of cutting-edge methodologies for hazard identification and risk assessment in the AM industry.
Collapse
Affiliation(s)
- Andi Alijagic
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Magnus Engwall
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Eva Särndahl
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Helen Karlsson
- Department of Health, Medicine and Caring Sciences, Occupational and Environmental Medicine Center in Linköping, Linköping University, Linköping, Sweden
| | - Alexander Hedbrant
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Lena Andersson
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Occupational and Environmental Medicine, Örebro University, Örebro, Sweden
| | - Patrik Karlsson
- Department of Mechanical Engineering, Örebro University, Örebro, Sweden
| | | | - Nikolai Scherbak
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | | | - Maria Larsson
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Alexander Persson
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
13
|
Martynova E, Rizvanov A, Urbanowicz RA, Khaiboullina S. Inflammasome Contribution to the Activation of Th1, Th2, and Th17 Immune Responses. Front Microbiol 2022; 13:851835. [PMID: 35369454 PMCID: PMC8969514 DOI: 10.3389/fmicb.2022.851835] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are cytosolic polyprotein complexes formed in response to various external and internal stimuli, including viral and bacterial antigens. The main product of the inflammasome is active caspase 1 which proteolytically cleaves, releasing functional interleukin-1 beta (IL-1β) and interleukin-18 (IL-18). These cytokines play a central role in shaping immune response to pathogens. In this review, we will focus on the mechanisms of inflammasome activation, as well as their role in development of Th1, Th2, and Th17 lymphocytes. The contribution of cytokines IL-1β, IL-18, and IL-33, products of activated inflammasomes, are summarized. Additionally, the role of cytokines released from tissue cells in promoting differentiation of lymphocyte populations is discussed.
Collapse
Affiliation(s)
| | | | - Richard A. Urbanowicz
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
14
|
Lee K, Lee J, Lee P, Jeon BC, Song MY, Kwak S, Lee J, Kim J, Kim D, Kim JH, Tesh VL, Lee M, Park S. Inhibition of O-GlcNAcylation protects from Shiga toxin-mediated cell injury and lethality in host. EMBO Mol Med 2022; 14:e14678. [PMID: 34842355 PMCID: PMC8749473 DOI: 10.15252/emmm.202114678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 11/09/2022] Open
Abstract
Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli (EHEC) are the major virulence factors responsible for hemorrhagic colitis, which can lead to life-threatening systemic complications including acute renal failure (hemolytic uremic syndrome) and neuropathy. Here, we report that O-GlcNAcylation, a type of post-translational modification, was acutely increased upon induction of endoplasmic reticulum (ER) stress in host cells by Stxs. Suppression of the abnormal Stx-mediated increase in O-GlcNAcylation effectively inhibited apoptotic and inflammatory responses in Stx-susceptible cells. The protective effect of O-GlcNAc inhibition for Stx-mediated pathogenic responses was also verified using three-dimensional (3D)-cultured spheroids or organoids mimicking the human kidney. Treatment with an O-GlcNAcylation inhibitor remarkably improved the major disease symptoms and survival rate for mice intraperitoneally injected with a lethal dose of Stx. In conclusion, this study elucidates O-GlcNAcylation-dependent pathogenic mechanisms of Stxs and demonstrates that inhibition of aberrant O-GlcNAcylation is a potential approach to treat Stx-mediated diseases.
Collapse
Affiliation(s)
- Kyung‐Soo Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Jieun Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Pureum Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Bong Chan Jeon
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
- Immunotherapy Convergence Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Min Yeong Song
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Sojung Kwak
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Jungwoon Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Jun‐Seob Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheonKorea
| | - Doo‐Jin Kim
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Ji Hyung Kim
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and ImmunologyCollege of MedicineTexas A&M UniversityBryanTXUSA
| | - Moo‐Seung Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Sung‐Kyun Park
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| |
Collapse
|
15
|
Farag VM, El-Shafei RA, Elkenany RM, Ali HS, Eladl AH. Antimicrobial, immunological and biochemical effects of florfenicol and garlic (Allium sativum) on rabbits infected with Escherichia coli serotype O55: H7. Vet Res Commun 2021; 46:363-376. [PMID: 34755272 DOI: 10.1007/s11259-021-09859-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 11/04/2021] [Indexed: 12/01/2022]
Abstract
Florfenicol (FFC) is a synthetic broad-spectrum antibiotic and garlic has a bactericidal action against coliforms. This study was carried out to compare the antimicrobial, immunological and biochemical effects of florfenicol and garlic, for their ability to treat enteropathogenic Escherichia coli serotype O55: H7 infection in rabbits. Four groups (G1-G4) were included. G1 group was the negative control; G2 group was the infected with a field-isolated strain of E. coli and untreated; G3 group was the infected+treated with FFC for 5 days; and G4 group was the infected+treated with garlic tablets for 14 days. The rabbits were observed for clinical signs, growth performance and mortality rates. Garlic-infused disks had a larger clear zone of inhibition than other antibiotic disks. Garlic treatment improved growth performance, biochemical parameters, and immunological response and reduced the fecal shedding and histopathological lesions in E. coli O55: H7 infected rabbits compared to the other groups. Colonization of E. coli more rapidly declined in G3 & G4 than in G2. Hepatic and intestinal gene expressions; tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were significantly elevated in G2 compared to the other groups, and their levels were elevated more in G3 than in G4. Serum interferon-gamma (IFN-γ) and phagocytic activity were significantly elevated in G4 compared to G3. G3 revealed macrocytic hypochromic anaemia that was confirmed histopathologically by moderate haematopoiesis of the bone marrow. In conclusion, garlic powder can reduce rabbit colibacillosis, like FFC, and can enhance the immune status of rabbits.
Collapse
Affiliation(s)
- Verginia M Farag
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Reham A El-Shafei
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, PO Box: 35516, Mansoura, Egypt.
| | - Rasha M Elkenany
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Hanaa S Ali
- Department of Pathology, Animal Health Research Institute, Mansoura branch, Agriculture Research Center, Giza, Egypt
| | - Abdelfattah H Eladl
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
Liu Y, Tian S, Thaker H, Dong M. Shiga Toxins: An Update on Host Factors and Biomedical Applications. Toxins (Basel) 2021; 13:222. [PMID: 33803852 PMCID: PMC8003205 DOI: 10.3390/toxins13030222] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/18/2022] Open
Abstract
Shiga toxins (Stxs) are classic bacterial toxins and major virulence factors of toxigenic Shigella dysenteriae and enterohemorrhagic Escherichia coli (EHEC). These toxins recognize a glycosphingolipid globotriaosylceramide (Gb3/CD77) as their receptor and inhibit protein synthesis in cells by cleaving 28S ribosomal RNA. They are the major cause of life-threatening complications such as hemolytic uremic syndrome (HUS), associated with severe cases of EHEC infection, which is the leading cause of acute kidney injury in children. The threat of Stxs is exacerbated by the lack of toxin inhibitors and effective treatment for HUS. Here, we briefly summarize the Stx structure, subtypes, in vitro and in vivo models, Gb3 expression and HUS, and then introduce recent studies using CRISPR-Cas9-mediated genome-wide screens to identify the host cell factors required for Stx action. We also summarize the latest progress in utilizing and engineering Stx components for biomedical applications.
Collapse
Affiliation(s)
- Yang Liu
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA; (S.T.); (H.T.)
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Songhai Tian
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA; (S.T.); (H.T.)
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Hatim Thaker
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA; (S.T.); (H.T.)
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Min Dong
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA; (S.T.); (H.T.)
- Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Li Z, Huang Z, Bai L. The P2X7 Receptor in Osteoarthritis. Front Cell Dev Biol 2021; 9:628330. [PMID: 33644066 PMCID: PMC7905059 DOI: 10.3389/fcell.2021.628330] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease. With the increasing aging population, the associated socio-economic costs are also increasing. Analgesia and surgery are the primary treatment options in late-stage OA, with drug treatment only possible in early prevention to improve patients' quality of life. The most important structural component of the joint is cartilage, consisting solely of chondrocytes. Instability in chondrocyte balance results in phenotypic changes and cell death. Therefore, cartilage degradation is a direct consequence of chondrocyte imbalance, resulting in the degradation of the extracellular matrix and the release of pro-inflammatory factors. These factors affect the occurrence and development of OA. The P2X7 receptor (P2X7R) belongs to the purinergic receptor family and is a non-selective cation channel gated by adenosine triphosphate. It mediates Na+, Ca2+ influx, and K+ efflux, participates in several inflammatory reactions, and plays an important role in the different mechanisms of cell death. However, the relationship between P2X7R-mediated cell death and the progression of OA requires investigation. In this review, we correlate potential links between P2X7R, cartilage degradation, and inflammatory factor release in OA. We specifically focus on inflammation, apoptosis, pyroptosis, and autophagy. Lastly, we discuss the therapeutic potential of P2X7R as a potential drug target for OA.
Collapse
Affiliation(s)
- Zihao Li
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyu Huang
- Foreign Languages College, Shanghai Normal University, Shanghai, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Song L, Xiao Y, Li X, Huang Y, Meng G, Ren Z. Activation of the Nlrp3 Inflammasome Contributes to Shiga Toxin-Induced Hemolytic Uremic Syndrome in a Mouse Model. Front Immunol 2021; 11:619096. [PMID: 33552083 PMCID: PMC7859089 DOI: 10.3389/fimmu.2020.619096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/03/2020] [Indexed: 11/13/2022] Open
Abstract
Objective To explore the role of the Nlrp3 inflammasome activation in the development of hemolytic uremic syndrome (HUS) induced by Stx2 and evaluate the efficacy of small molecule Nlrp3 inhibitors in preventing the HUS. Methods Peritoneal macrophages (PMs) isolated from wild-type (WT) C57BL/6J mice and gene knockout mice (Nlrc4 -/-, Aim2 -/-, and Nlrp3 -/-) were treated with Stx2 in vitro and their IL-1β releases were measured. WT mice and Nlrp3 -/- mice were also treated with Stx2 in vivo by injection, and the biochemical indices (serum IL-1β, creatinine [CRE] and blood urea nitrogen [BUN]), renal injury, and animal survival were compared. To evaluate the effect of the Nlrp3 inhibitors in preventing HUS, WT mice were pretreated with different Nlrp3 inhibitors (MCC950, CY-09, Oridonin) before Stx2 treatment, and their biochemical indices and survival were compared with the WT mice without inhibitor pretreatment. Results When PMs were stimulated by Stx2 in vitro, IL-1β release in Nlrp3 -/- PMs was significantly lower compared to the other PMs. The Nlrp3 -/- mice treated by Stx2 in vivo, showed lower levels of the biochemical indices, alleviated renal injuries, and increased survival rate. When the WT mice were pretreated with the Nlrp3 inhibitors, both the biochemical indices and survival were significantly improved compared to those without inhibitor pretreatment, with Oridonin being most potent. Conclusion Nlrp3 inflammasome activation plays a vital role in the HUS development when mice are challenged by Stx2, and Oridonin is effective in preventing HUS.
Collapse
Affiliation(s)
- Liqiong Song
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuchun Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Xianping Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanming Huang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Guangxun Meng
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, Chinese Academy of Sciences, Institute Pasteur of Shanghai, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhihong Ren
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
19
|
Jing W, Lo Pilato J, Kay C, Man SM. Activation mechanisms of inflammasomes by bacterial toxins. Cell Microbiol 2021; 23:e13309. [PMID: 33426791 DOI: 10.1111/cmi.13309] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/16/2020] [Accepted: 12/12/2020] [Indexed: 12/13/2022]
Abstract
Inflammasomes are cytosolic innate immune complexes, which assemble in mammalian cells in response to microbial components and endogenous danger signals. A major family of inflammasome activators is bacterial toxins. Inflammasome sensor proteins, such as the nucleotide-binding oligomerisation domain-like receptor (NLR) family members NLRP1b and NLRP3, and the tripartite motif family member Pyrin+ efflux triggered by pore-forming toxins or by other toxin-induced homeostasis-altering events such as lysosomal rupture. Pyrin senses perturbation of host cell functions induced by certain enzymatic toxins resulting in impairment of RhoA GTPase activity. Assembly of the inflammasome complex activates the cysteine protease caspase-1, leading to the proteolytic cleavage of the proinflammatory cytokines IL-1β and IL-18, and the pore-forming protein gasdermin D causing pyroptosis. In this review, we discuss the latest progress in our understanding on the activation mechanisms of inflammasome complexes by bacterial toxins and effector proteins and explore avenues for future research into the relationships between inflammasomes and bacterial toxins.
Collapse
Affiliation(s)
- Weidong Jing
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Jordan Lo Pilato
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Callum Kay
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Si Ming Man
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
20
|
Havira MS, Ta A, Kumari P, Wang C, Russo AJ, Ruan J, Rathinam VA, Vanaja SK. Shiga toxin suppresses noncanonical inflammasome responses to cytosolic LPS. Sci Immunol 2020; 5:5/53/eabc0217. [PMID: 33246946 DOI: 10.1126/sciimmunol.abc0217] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/02/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory caspase-dependent cytosolic lipopolysaccharide (LPS) sensing is a critical arm of host defense against bacteria. How pathogens overcome this pathway to establish infections is largely unknown. Enterohemorrhagic Escherichia coli (EHEC) is a clinically important human pathogen causing hemorrhagic colitis and hemolytic uremic syndrome. We found that a bacteriophage-encoded virulence factor of EHEC, Shiga toxin (Stx), suppresses caspase-11-mediated activation of the cytosolic LPS sensing pathway. Stx was essential and sufficient to inhibit pyroptosis and interleukin-1 (IL-1) responses elicited specifically by cytosolic LPS. The catalytic activity of Stx was necessary for suppression of inflammasome responses. Stx impairment of inflammasome responses to cytosolic LPS occurs at the level of gasdermin D activation. Stx also suppresses inflammasome responses in vivo after LPS challenge and bacterial infection. Overall, this study assigns a previously undescribed inflammasome-subversive function to a well-known bacterial toxin, Stx, and reveals a new phage protein-based pathogen blockade of cytosolic immune surveillance.
Collapse
Affiliation(s)
- Morena S Havira
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Atri Ta
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Puja Kumari
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Chengliang Wang
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ashley J Russo
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Jianbin Ruan
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, UConn Health School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA.
| |
Collapse
|
21
|
Lee MS, Yoon JW, Tesh VL. Editorial: Recent Advances in Understanding the Pathogenesis of Shiga Toxin-Producing Shigella and Escherichia coli. Front Cell Infect Microbiol 2020; 10:620703. [PMID: 33324585 PMCID: PMC7726016 DOI: 10.3389/fcimb.2020.620703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jang Won Yoon
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, South Korea
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College of Medicine, Bryan, TX, United States
| |
Collapse
|
22
|
Zhou SY, Cui GZ, Yan XL, Wang X, Qu Y, Guo ZN, Jin H. Mechanism of Ferroptosis and Its Relationships With Other Types of Programmed Cell Death: Insights for Potential Interventions After Intracerebral Hemorrhage. Front Neurosci 2020; 14:589042. [PMID: 33281547 PMCID: PMC7691292 DOI: 10.3389/fnins.2020.589042] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a fatal cerebrovascular disease with high morbidity and mortality, for which no effective therapies are currently available. Brain tissue damage caused by ICH is mediated by a newly identified form of non-apoptotic programmed cell death, called ferroptosis. Ferroptosis is characterized by the iron-induced accumulation of lipid reactive oxygen species (ROS), leading to intracellular oxidative stress. Lipid ROS cause damage to nucleic acids, proteins, and cell membranes, eventually resulting in ferroptosis. Numerous biological processes are involved in ferroptosis, including iron metabolism, lipid peroxidation, and glutathione biosynthesis; therefore, iron chelators, lipophilic antioxidants, and other specific inhibitors can suppress ferroptosis, suggesting that these modulators are beneficial for treating brain injury due to ICH. Accumulating evidence indicates that ferroptosis differs from other types of programmed cell death, such as necroptosis, apoptosis, oxytosis, and pyroptosis, in terms of ultrastructural characteristics, signaling pathways, and outcomes. Although several studies have emphasized the importance of ferroptosis due to ICH, the detailed mechanism underlying ferroptosis remains unclear. This review summarizes the available evidence on the mechanism underlying ferroptosis and its relationship with other types of cell death, with the aim to identify therapeutic targets and potential interventions for ICH.
Collapse
Affiliation(s)
- Sheng-Yu Zhou
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Guo-Zhen Cui
- Department of Hepatology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Qu
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Zhen-Ni Guo
- Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Hang Jin
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Liu L, Song L, Deng R, Lan R, Jin W, Tran Van Nhieu G, Cao H, Liu Q, Xiao Y, Li X, Meng G, Ren Z. Citrobacter freundii Activation of NLRP3 Inflammasome via the Type VI Secretion System. J Infect Dis 2020; 223:2174-2185. [PMID: 33151309 DOI: 10.1093/infdis/jiaa692] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022] Open
Abstract
Citrobacter freundii is a significant cause of human infections, responsible for food poisoning, diarrhea, and urinary tract infections. We previously identified a highly cytotoxic and adhesive C. freundii strain CF74 expressing a type VI secretion system (T6SS). In this study, we showed that in mice-derived macrophages, C. freundii CF74 activated the Nucleotide Oligomerization Domain -Like Receptor Family, Pyrin Domain Containing 3(NLRP3) inflammasomes in a T6SS-dependent manner. The C. freundii T6SS activated the inflammasomes mainly through caspase 1 and mediated pyroptosis of macrophages by releasing the cleaved gasdermin-N domain. The CF74 T6SS was required for flagellin-induced interleukin 1β release by macrophages. We further show that the T6SS tail component and effector, hemolysin co-regulation protein-2 (Hcp-2), was necessary and sufficient to trigger NLRP3 inflammasome activation. In vivo, the T6SS played a key role in mediating interleukin 1β secretion and the survival of mice during C. freundii infection in mice. These findings provide novel insights into the role of T6SS in the pathogenesis of C. freundii.
Collapse
Affiliation(s)
- Liyun Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Liqiong Song
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Rong Deng
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Wenjie Jin
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Guy Tran Van Nhieu
- Calcium Signaling and Microbial Infections, Inserm U1282, Laboratoire de Biologie et Pharmacologie Appliquée, UMR 8113, Ecole Normale Supérieure Paris Saclay, Gif-sur-Yvette, France
| | - Huifang Cao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuchun Xiao
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Xianping Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Guangxun Meng
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China
| | - Zhihong Ren
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018RU010), Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Harrison LM, Lacher DW, Mammel MK, Leonard SR. Comparative Transcriptomics of Shiga Toxin-Producing and Commensal Escherichia coli and Cytokine Responses in Colonic Epithelial Cell Culture Infections. Front Cell Infect Microbiol 2020; 10:575630. [PMID: 33194815 PMCID: PMC7649339 DOI: 10.3389/fcimb.2020.575630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Ingestion of Shiga toxin-producing Escherichia coli (STEC) can result in a range of illness severity from asymptomatic to hemorrhagic colitis and death; thus risk assessment of STEC strains for human pathogenicity is important in the area of food safety. Illness severity depends in part on the combination of virulence genes carried in the genome, which can vary between strains even of identical serotype. To better understand how core genes are regulated differently among strains and to identify possible novel STEC virulence gene candidates that could be added to the risk assessment repertoire, we used comparative transcriptomics to investigate global gene expression differences between two STEC strains associated with severe illness and a commensal E. coli strain during in vitro intestinal epithelial cell (IEC) infections. Additionally, we compared a wide array of concomitant cytokine levels produced by the IECs. The cytokine expression levels were examined for a pattern representing STEC pathogenicity; however, while one STEC strain appeared to elicit a proinflammatory response, infection by the other strain produced a pattern comparable to the commensal E. coli. This result may be explained by the significant differences in gene content and expression observed between the STEC strains. RNA-Seq analysis revealed considerable disparity in expression of genes in the arginine and tryptophan biosynthesis/import pathways between the STEC strains and the commensal E. coli strain, highlighting the important role some amino acids play in STEC colonization and survival. Contrasting differential expression patterns were observed for genes involved in respiration among the three strains suggesting that metabolic diversity is a strategy utilized to compete with resident microflora for successful colonization. Similar temporal expression results for known and putative virulence genes were observed in the STEC strains, revealing strategies used for survival prior to and after initial adherence to IECs. Additionally, three genes encoding hypothetical proteins located in mobile genetic elements were, after interrogation of a large set of E. coli genomes, determined to likely represent novel STEC virulence factors.
Collapse
Affiliation(s)
- Lisa M Harrison
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - David W Lacher
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - Mark K Mammel
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - Susan R Leonard
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| |
Collapse
|
25
|
Lee KS, Lee J, Lee P, Kim CU, Kim DJ, Jeong YJ, Park YJ, Tesh VL, Lee MS. Exosomes released from Shiga toxin 2a-treated human macrophages modulate inflammatory responses and induce cell death in toxin receptor expressing human cells. Cell Microbiol 2020; 22:e13249. [PMID: 32772454 DOI: 10.1111/cmi.13249] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/02/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
Shiga toxins (Stxs) produced by Stx-producing Escherichia coli are the primarily virulence factors of hemolytic uremic syndrome and central nervous system (CNS) impairment. Although the precise mechanisms of toxin dissemination remain unclear, Stxs bind to extracellular vesicles (EVs). Exosomes, a subset of EVs, may play a key role in Stx-mediated renal injury. To test this hypothesis, we isolated exosomes from monocyte-derived macrophages in the presence of Stx2a or Stx2 toxoids. Macrophage-like differentiated THP-1 cells treated with Stxs secreted Stx-associated exosomes (Stx-Exo) of 90-130 nm in diameter, which induced cytotoxicity in recipient cells in a toxin receptor globotriaosylceramide (Gb3 )-dependent manner. Stx2-Exo engulfed by Gb3 -positive cells were translocated to the endoplasmic reticulum in the human proximal tubule epithelial cell line HK-2. Stx2-Exo contained pro-inflammatory cytokine mRNAs and proteins and induced more severe inflammation than purified Stx2a accompanied by greater death of target cells such as human renal or retinal pigment epithelial cells. Blockade of exosome biogenesis using the pharmacological inhibitor GW4869 reduced Stx2-Exo-mediated human renal cell death. Stx2-Exo isolated from human primary monocyte-derived macrophages activated caspase 3/7 and resulted in significant cell death in primary human renal cortical epithelial cells. Based on these results, we speculate that Stx-containing exosomes derived from macrophages may exacerbate cytotoxicity and inflammation and trigger cell death in toxin-sensitive cells. Therapeutic interventions targeting Stx-containing exosomes may prevent or ameliorate Stx-mediated acute vascular dysfunction.
Collapse
Affiliation(s)
- Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jieun Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Pureum Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Young-Jun Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College of Medicine, Bryan, Texas, USA
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| |
Collapse
|
26
|
Mercatelli D, Bortolotti M, Giorgi FM. Transcriptional network inference and master regulator analysis of the response to ribosome-inactivating proteins in leukemia cells. Toxicology 2020; 441:152531. [PMID: 32593706 DOI: 10.1016/j.tox.2020.152531] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022]
Abstract
Gene-regulatory networks reconstruction has become a very popular approach in applied biology to infer and dissect functional interactions of Transcription Factors (TFs) driving a defined phenotypic state, termed as Master Regulators (MRs). In the present work, cutting-edge bioinformatic methods were applied to re-analyze experimental data on leukemia cells (human myelogenous leukemia cell line THP-1 and acute myeloid leukemia MOLM-13 cells) treated for 6 h with two different Ribosome-Inactivating Proteins (RIPs), namely Shiga toxin type 1 (400 ng/mL) produced by Escherichia coli strains and the plant toxin stenodactylin (60 ng/mL), purified from the caudex of Adenia stenodactyla Harms. This analysis allowed us to identify the common early transcriptional response to 28S rRNA damage based on gene-regulatory network inference and Master Regulator Analysis (MRA). Both toxins induce a common response at 6 h which involves inflammatory mediators triggered by AP-1 family transcriptional factors and ATF3 in leukemia cells. We describe for the first time the involvement of MAFF, KLF2 and KLF6 in regulating RIP-induced apoptotic cell death, while receptor-mediated downstream signaling through ANXA1 and TLR4 is suggested for both toxins.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Italy.
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Italy.
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Italy.
| |
Collapse
|
27
|
Gerbino E, Ghibaudo F, Tymczyszyn EE, Gomez-Zavaglia A, Hugo AA. Probiotics, Galacto-oligosaccharides, and zinc antagonize biological effects of enterohaemorrhagic Escherichia coli on cultured cells and brine shrimp model. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2020.109435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Molecular Biology of Escherichia Coli Shiga Toxins' Effects on Mammalian Cells. Toxins (Basel) 2020; 12:toxins12050345. [PMID: 32456125 PMCID: PMC7290813 DOI: 10.3390/toxins12050345] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Shiga toxins (Stxs), syn. Vero(cyto)toxins, are potent bacterial exotoxins and the principal virulence factor of enterohemorrhagic Escherichia coli (EHEC), a subset of Shiga toxin-producing E. coli (STEC). EHEC strains, e.g., strains of serovars O157:H7 and O104:H4, may cause individual cases as well as large outbreaks of life-threatening diseases in humans. Stxs primarily exert a ribotoxic activity in the eukaryotic target cells of the mammalian host resulting in rapid protein synthesis inhibition and cell death. Damage of endothelial cells in the kidneys and the central nervous system by Stxs is central in the pathogenesis of hemolytic uremic syndrome (HUS) in humans and edema disease in pigs. Probably even more important, the toxins also are capable of modulating a plethora of essential cellular functions, which eventually disturb intercellular communication. The review aims at providing a comprehensive overview of the current knowledge of the time course and the consecutive steps of Stx/cell interactions at the molecular level. Intervention measures deduced from an in-depth understanding of this molecular interplay may foster our basic understanding of cellular biology and microbial pathogenesis and pave the way to the creation of host-directed active compounds to mitigate the pathological conditions of STEC infections in the mammalian body.
Collapse
|
29
|
Involvement of NF-κB1 and the Non-Canonical NF-κB Signaling Pathway in the Pathogenesis of Acute Kidney Injury in Shiga-Toxin-2-Induced Hemolytic-Uremic Syndrome in Mice. Shock 2020; 56:573-581. [PMID: 32433206 DOI: 10.1097/shk.0000000000001558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The hemolytic-uremic syndrome (HUS) is a thrombotic microangiopathy which can occur as a severe systemic complication after an infection with Shiga-toxin-(Stx)-producing Escherichia coli (STEC). Elevated levels of proinflammatory cytokines associated with the classical NF-κB signaling pathway were detected in the urine of HUS patients. Thus, we hypothesize that the immune response of the infected organism triggered by Stx can affect the kidneys and contributes to acute kidney injury. Hitherto the role of the classical and non-canonical NF-κB signaling pathway in HUS has not been evaluated systematically in vivo. We aimed to investigate in a murine model of Shiga toxin-induced HUS-like disease, whether one or both pathways are involved in the renal pathology in HUS. In kidneys of mice subjected to Stx or sham-treated mice, protein or gene expression analyses were performed to assess the 1) expression of receptors activating the classical and non-canonical pathway, such as Fn14 and CD40 2) levels of NF-κB1/RelA and NF-κB2/RelB including its upstream signaling proteins and 3) expression of cytokines as target molecules of both pathways. In line with a higher expression of Fn14 and CD40, we detected an enhanced translocation of NF-κB1 and RelA as well as NF-κB2 and RelB into the nucleus accompanied by an increased gene expression of the NF-κB1-target cytokines Ccl20, Cxcl2, Ccl2, Cxcl1, IL-6, TNF-α, Cxcl10 and Ccl5, indicating an activation of the classical and non-canonical NF-κB pathway. Thereby, we provide, for the first time, in vivo evidence for an involvement of both NF-κB signaling pathways in renal pathophysiology of STEC-HUS.
Collapse
|
30
|
Mercatelli D, Bortolotti M, Andresen V, Sulen A, Polito L, Gjertsen BT, Bolognesi A. Early Response to the Plant Toxin Stenodactylin in Acute Myeloid Leukemia Cells Involves Inflammatory and Apoptotic Signaling. Front Pharmacol 2020; 11:630. [PMID: 32457623 PMCID: PMC7226368 DOI: 10.3389/fphar.2020.00630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
Stenodactylin, a highly toxic type 2 ribosome-inactivating protein purified from the caudex of Adenia stenodactyla Harms, is a potential anticancer drug candidate. Previous studies demonstrated that stenodactylin induces apoptosis and necroptosis in treated cells, involving the production of reactive oxygen species. We analyzed the effect of stenodactylin on Raji and Ramos (Human Burkitt’s lymphoma cells) and MOLM-13 (acute myeloid leukemia cells). Moreover, we focused on the early events in MOLM-13 cells that characterize the cellular response to the toxin by whole-genome microarray analysis of gene expression. Treatment with stenodactylin induced the depurination of 28S rRNA within 4 h and increased the phosphorylation of p38 and JNK. A time-dependent activation of caspase 1, 2, 8, 9, 3/7 was also observed. Genome-wide gene expression microarray analysis revealed early changes in the expression of genes involved in the regulation of cell death, inflammation and stress response. After 4 h, a significant increase of transcript level was detectable for ATF3, BTG2, DUSP1, EGR1, and JUN. Increased upstream JUN signaling was also confirmed at protein level. The early response to stenodactylin treatment involves inflammatory and apoptotic signaling compatible with the activation of multiple cell death pathways. Because of the above described properties toward acute myeloid leukemia cells, stenodactylin may be a promising candidate for the design of new immunoconjugates for experimental cancer treatment.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology-FaBiT, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Vibeke Andresen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hematology Section, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
| | - André Sulen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Bjørn Tore Gjertsen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hematology Section, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
| | - Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
31
|
Sanchez‐Garrido J, Slater SL, Clements A, Shenoy AR, Frankel G. Vying for the control of inflammasomes: The cytosolic frontier of enteric bacterial pathogen-host interactions. Cell Microbiol 2020; 22:e13184. [PMID: 32185892 PMCID: PMC7154749 DOI: 10.1111/cmi.13184] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/13/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022]
Abstract
Enteric pathogen-host interactions occur at multiple interfaces, including the intestinal epithelium and deeper organs of the immune system. Microbial ligands and activities are detected by host sensors that elicit a range of immune responses. Membrane-bound toll-like receptors and cytosolic inflammasome pathways are key signal transducers that trigger the production of pro-inflammatory molecules, such as cytokines and chemokines, and regulate cell death in response to infection. In recent years, the inflammasomes have emerged as a key frontier in the tussle between bacterial pathogens and the host. Inflammasomes are complexes that activate caspase-1 and are regulated by related caspases, such as caspase-11, -4, -5 and -8. Importantly, enteric bacterial pathogens can actively engage or evade inflammasome signalling systems. Extracellular, vacuolar and cytosolic bacteria have developed divergent strategies to subvert inflammasomes. While some pathogens take advantage of inflammasome activation (e.g. Listeria monocytogenes, Helicobacter pylori), others (e.g. E. coli, Salmonella, Shigella, Yersinia sp.) deploy a range of virulence factors, mainly type 3 secretion system effectors, that subvert or inhibit inflammasomes. In this review we focus on inflammasome pathways and their immune functions, and discuss how enteric bacterial pathogens interact with them. These studies have not only shed light on inflammasome-mediated immunity, but also the exciting area of mammalian cytosolic immune surveillance.
Collapse
Affiliation(s)
| | | | | | - Avinash R. Shenoy
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology & InfectionImperial College LondonLondonUK
| | - Gad Frankel
- Department of Life SciencesImperial College LondonLondonUK
| |
Collapse
|
32
|
Role of Shiga Toxins in Cytotoxicity and Immunomodulatory Effects of Escherichia coli O157:H7 during Host-Bacterial Interactions in vitro. Toxins (Basel) 2020; 12:toxins12010048. [PMID: 31947665 PMCID: PMC7020462 DOI: 10.3390/toxins12010048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/19/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) strains are food-borne pathogens that can cause different clinical conditions. Shiga toxin 2a and/or 2c (Stx2)-producing E. coli O157:H7 is the serotype most frequently associated with severe human disease. In this work we analyzed the hypothesis that host cells participate in Stx2 production, cell damage, and inflammation during EHEC infection. With this aim, macrophage-differentiated THP-1 cells and the intestinal epithelial cell line HCT-8 were incubated with E. coli O157:H7. A time course analysis of cellular and bacterial survival, Stx2 production, stx2 transcription, and cytokine secretion were analyzed in both human cell lines. We demonstrated that macrophages are able to internalize and kill EHEC. Simultaneously, Stx2 produced by internalized bacteria played a major role in macrophage death. In contrast, HCT-8 cells were completely resistant to EHEC infection. Besides, macrophages and HCT-8 infected cells produce IL-1β and IL-8 inflammatory cytokines, respectively. At the same time, bacterial stx2-specific transcripts were detected only in macrophages after EHEC infection. The interplay between bacteria and host cells led to Stx production, triggering of inflammatory response and cell damage, all of which could contribute to a severe outcome after EHEC infections.
Collapse
|
33
|
Platnich JM, Chung H, Lau A, Sandall CF, Bondzi-Simpson A, Chen HM, Komada T, Trotman-Grant AC, Brandelli JR, Chun J, Beck PL, Philpott DJ, Girardin SE, Ho M, Johnson RP, MacDonald JA, Armstrong GD, Muruve DA. Shiga Toxin/Lipopolysaccharide Activates Caspase-4 and Gasdermin D to Trigger Mitochondrial Reactive Oxygen Species Upstream of the NLRP3 Inflammasome. Cell Rep 2019; 25:1525-1536.e7. [PMID: 30404007 DOI: 10.1016/j.celrep.2018.09.071] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 06/04/2018] [Accepted: 09/21/2018] [Indexed: 01/11/2023] Open
Abstract
The non-canonical caspase-4 and canonical NLRP3 inflammasomes are both activated by intracellular lipopolysaccharide (LPS), but the crosstalk between these two pathways remains unclear. Shiga toxin 2 (Stx2)/LPS complex, from pathogenic enterohemorrhagic Escherichia coli, activates caspase-4, gasdermin D (GSDMD), and the NLRP3 inflammasome in human THP-1 macrophages, but not mouse macrophages that lack the Stx receptor CD77. Stx2/LPS-mediated IL-1β secretion and pyroptosis are dependent on mitochondrial reactive oxygen species (ROS) downstream of the non-canonical caspase-4 inflammasome and cleaved GSDMD, which is enriched at the mitochondria. Blockade of caspase-4 activation and ROS generation as well as GSDMD deficiency significantly reduces Stx2/LPS-induced IL-1β production and pyroptosis. The NLRP3 inflammasome plays a significant role in amplifying Stx2/LPS-induced GSDMD cleavage and pyroptosis, with significant reduction of these responses in NLRP3-deficient THP-1 cells. Together, these data show that Stx2/LPS complex activates the non-canonical inflammasome and mitochondrial ROS upstream of the NLRP3 inflammasome to promote cytokine maturation and pyroptosis.
Collapse
Affiliation(s)
- Jaye M Platnich
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Hyunjae Chung
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Arthur Lau
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Christina F Sandall
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Adom Bondzi-Simpson
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Huey-Miin Chen
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Takanori Komada
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | | | - Jeremy R Brandelli
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Justin Chun
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Paul L Beck
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Stephen E Girardin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - May Ho
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Roger P Johnson
- Public Health Agency of Canada, National Microbiology Laboratory, Guelph, ON, Canada
| | - Justin A MacDonald
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Glen D Armstrong
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Daniel A Muruve
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
34
|
Winsor N, Krustev C, Bruce J, Philpott DJ, Girardin SE. Canonical and noncanonical inflammasomes in intestinal epithelial cells. Cell Microbiol 2019; 21:e13079. [PMID: 31265745 DOI: 10.1111/cmi.13079] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
Inflammasomes are cytosolic, multimeric protein complexes capable of activating pro-inflammatory cytokines such as IL-1β and IL-18, which play a key role in host defence. Inflammasome components are highly expressed in the intestinal epithelium. In recent years, studies have begun to demonstrate that epithelial-intrinsic inflammasomes play a critical role in regulating epithelial homeostasis, both by defending the epithelium from pathogenic insult and through the regulation of the mucosal environment. However, the majority of research regarding inflammasome activation has focused on professional immune cells, such as macrophages. Here, we present an overview of the current understanding of inflammasome function in epithelial cells and at mucosal surfaces and, in particular, in the intestine.
Collapse
Affiliation(s)
- Nathaniel Winsor
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Christian Krustev
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Bruce
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen E Girardin
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Eladl AH, Farag VM, El-Shafei RA, Elkenany RM, Elsayed MM, Mona MM, Ali HS, Saif MA. Effect of colibacillosis on the immune response to a rabbit viral haemorrhagic disease vaccine. Vet Microbiol 2019; 238:108429. [PMID: 31648721 DOI: 10.1016/j.vetmic.2019.108429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 01/03/2023]
Abstract
Viral haemorrhagic disease (VHD) and colibacillosis are common diseases in rabbits that cause economic losses worldwide. The effect of colibacillosis on the immune response of vaccinated rabbits against rabbit haemorrhagic disease virus (RHDV) was studied. Four groups (G1-G4) were included. G1 was the negative control group; G2 was the RHDV vaccine group; G3 was the E. coli-infected group; and G4 was the E. coli-infected + RHDV vaccine group. The E. coli infection and RHDV vaccination were simultaneously performed, with another previous infection, 3 days before vaccination. At 28 days post-vaccination (PV), the rabbits (G2-G4) were challenged intramuscularly with 0.5 ml of RHDV at a dose of 103 50% median lethal dose (LD50)/rabbit. The rabbits were observed for clinical signs, body weight gain and mortality rates. Tissue, blood, serum, and faecal samples and rectal swabs were collected at 3, 5, 7, 14, 21 and 28 days PV. Significant clinical signs and mortality and a decrease in BW were observed in the infected + RHDV vaccine group. On the 3rd day post-infection (PI), compared with all the other groups, the vaccinated group (G2) had significantly upregulated hepatic tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels; however, the infected + RHDV vaccine group had significantly higher intestinal levels of TNF-α and IL-6 than the other groups. Furthermore, E. coli infection in vaccinated rabbits led to immunosuppression, as shown by significant decreases (P < 0.05) in heterophil phagocytic activity, the CD4+/CD8+ ratio, and HI antibody responses to RHDV and a significant increase in the heterophil to lymphocyte (H/L) ratio. In conclusion, colibacillosis leads to immunosuppression involving a shift in the equilibrium of cytokines and reduced weight gain and mortality in vaccinated rabbits and could be a contributing factor in RHDV vaccination failure in rabbit farming.
Collapse
Affiliation(s)
- Abdelfattah H Eladl
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| | - Verginia M Farag
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Reham A El-Shafei
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Rasha M Elkenany
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mona M Elsayed
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Marwa M Mona
- Department of Medical Biochemistry, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Hanaa S Ali
- Department of Pathology, Animal Health Research Institute, Mansoura branch, Egypt
| | - Mohamed A Saif
- Researcher of Virology, Reference Laboratory of Quality Control of Poultry Production (Gamasa)- Animal Health Research Institute, Egypt
| |
Collapse
|
36
|
The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int J Mol Sci 2019; 20:ijms20133328. [PMID: 31284572 PMCID: PMC6651423 DOI: 10.3390/ijms20133328] [Citation(s) in RCA: 2070] [Impact Index Per Article: 345.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023] Open
Abstract
The NLRP3 inflammasome is a critical component of the innate immune system that mediates caspase-1 activation and the secretion of proinflammatory cytokines IL-1β/IL-18 in response to microbial infection and cellular damage. However, the aberrant activation of the NLRP3 inflammasome has been linked with several inflammatory disorders, which include cryopyrin-associated periodic syndromes, Alzheimer's disease, diabetes, and atherosclerosis. The NLRP3 inflammasome is activated by diverse stimuli, and multiple molecular and cellular events, including ionic flux, mitochondrial dysfunction, and the production of reactive oxygen species, and lysosomal damage have been shown to trigger its activation. How NLRP3 responds to those signaling events and initiates the assembly of the NLRP3 inflammasome is not fully understood. In this review, we summarize our current understanding of the mechanisms of NLRP3 inflammasome activation by multiple signaling events, and its regulation by post-translational modifications and interacting partners of NLRP3.
Collapse
|
37
|
Üçeyler N, Urlaub D, Mayer C, Uehlein S, Held M, Sommer C. Tumor necrosis factor-α links heat and inflammation with Fabry pain. Mol Genet Metab 2019; 127:200-206. [PMID: 31221509 DOI: 10.1016/j.ymgme.2019.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/08/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023]
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder associated with pain triggered by heat or febrile infections. We modelled this condition by measuring the cytokine expression of peripheral blood mononuclear cells (PBMC) from FD patients in vitro upon stimulation with heat and lipopolysaccharide (LPS). We enrolled 67 FD patients and 37 healthy controls. We isolated PBMC, assessed their gene expression of selected pro- and anti-inflammatory cytokines, incubated them with heat, LPS, globotriaosylceramide (Gb3), and tumor necrosis factor-α (TNF), and measured TNF secretion in the supernatant and intracellular Gb3 accumulation, respectively. We found increased TNF, interleukin (IL-)1β, and toll-like receptor 4 (TLR4) gene expression in FD men (p < .05 to p < .01). TNF and IL-10 were higher, and IL-4 was lower in the subgroup of FD men with pain compared to controls (p < .05 to p < .01). Hereby, TNF was only increased in FD men with pain and classical mutations (p < .05) compared to those without pain. PBMC from FD patients secreted more TNF upon stimulation with LPS (p < .01) than control PBMC. Incubation with Gb3 and an additional α-galactosidase A inhibitor did not further increase TNF secretion, but incubation with TNF greatly increased the Gb3 load in FD PBMC compared to controls (p < .01). Also, LPS incubation and heat challenge (40 °C) increased Gb3 accumulation in PBMC of patients compared to baseline (p < .05 each), while no alterations were observed in control PBMC. Our data show that TNF holds a crucial role in the pathophysiology of FD associated pain, which may open a novel perspective for analgesic treatment in FD pain.
Collapse
Affiliation(s)
- Nurcan Üçeyler
- Department of Neurology, University of Würzburg, Germany; Würzburg Fabry Center for Interdisciplinary Therapy (FAZIT), University of Würzburg, Germany.
| | - Daniela Urlaub
- Department of Neurology, University of Würzburg, Germany
| | | | | | - Melissa Held
- Department of Neurology, University of Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University of Würzburg, Germany; Würzburg Fabry Center for Interdisciplinary Therapy (FAZIT), University of Würzburg, Germany
| |
Collapse
|
38
|
Lee MS, Tesh VL. Roles of Shiga Toxins in Immunopathology. Toxins (Basel) 2019; 11:E212. [PMID: 30970547 PMCID: PMC6521259 DOI: 10.3390/toxins11040212] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/20/2022] Open
Abstract
Shigella species and Shiga toxin-producing Escherichia coli (STEC) are agents of bloody diarrhea that may progress to potentially lethal complications such as diarrhea-associated hemolytic uremic syndrome (D+HUS) and neurological disorders. The bacteria share the ability to produce virulence factors called Shiga toxins (Stxs). Research over the past two decades has identified Stxs as multifunctional toxins capable of inducing cell stress responses in addition to their canonical ribotoxic function inhibiting protein synthesis. Notably, Stxs are not only potent inducers of cell death, but also activate innate immune responses that may lead to inflammation, and these effects may increase the severity of organ injury in patients infected with Stx-producing bacteria. In the intestines, kidneys, and central nervous system, excessive or uncontrolled host innate and cellular immune responses triggered by Stxs may result in sensitization of cells to toxin mediated damage, leading to immunopathology and increased morbidity and mortality in animal models (including primates) and human patients. Here, we review studies describing Stx-induced innate immune responses that may be associated with tissue damage, inflammation, and complement activation. We speculate on how these processes may contribute to immunopathological responses to the toxins.
Collapse
Affiliation(s)
- Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea.
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
39
|
Sahin E, Bektur E, Burukoglu Donmez D, Baycu C, Can OD, Sahinturk V. Mirtazapine suppresses sterile inflammation through NLRP3-inflammasome in diabetic rat kidney. Acta Histochem 2019; 121:289-296. [PMID: 30711241 DOI: 10.1016/j.acthis.2019.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 01/25/2019] [Accepted: 01/25/2019] [Indexed: 12/21/2022]
Abstract
AIM The aim of this study was to investigate the effects of mirtazapine, which is anti-oxidative and antidepressant agent, on the kidney damage caused by diabetes mellitus. MATERIALS AND METHODS The rats were randomly divided into three groups (n = 7 animals in each group). The group I rats served as control and they received 0.1 mol/L of citric acid buffer (pH = 4.5) as vehicle. The rats in the group II (DM group) and III (DM + Mirtazapine-treated group) were treated intraperitoneally with a single dose of 55 mg/kg streptozotocin dissolved in 0.1 mol/L of citric acid buffer. Group III rats were also received 20 mg/kg/day of mirtazapine for 2 weeks. At the end of the experiment, the rats were sacrificed. Then, the kidneys were excised and prepared for microscopical examination. caspase-1 and NLRP3 proteins were examined using immunohistochemistry and western blotting. The TUNEL assay for apoptosis and ELISA assay for IL-1β were performed. RESULTS Histological examination showed that mirtazapine administration has an ameliorative effect on DM-induced kidney damage. Immunohistochemical and western blot analyses showed that NLRP3 and caspase-1 expressions were increased in the DM group according to the control group and the mirtazapine administration decreased these expressions. The intraglomerular and tubular TUNEL-positive cells were numerous in the DM group compared to the mirtazapine-treated group. The level of IL-1β was highest in the DM group, and decreased significantly in the mirtazapine-treated group. CONCLUSION In this study, 20 mg/kg/day mirtazapine administration for 2 weeks reduced NLRP3 and caspase-1 expressions and IL-1β level in the diabetic rat kidneys. These results suggesting that mirtazapine may be useful in the treatment of DM and other metabolic diseases. Advanced molecular studies are needed to elucidate the exact effects of mirtazapine on NLRP3 inflammasome.
Collapse
|
40
|
Shiga Toxin Type 1a (Stx1a) Reduces the Toxicity of the More Potent Stx2a In Vivo and In Vitro. Infect Immun 2019; 87:IAI.00787-18. [PMID: 30670557 DOI: 10.1128/iai.00787-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022] Open
Abstract
Shiga toxin (Stx)-producing Escherichia coli (STEC) causes foodborne outbreaks of bloody diarrhea. There are two major types of immunologically distinct Stxs: Stx1a and Stx2a. Stx1a is more cytotoxic to Vero cells than Stx2a, but Stx2a has a lower 50% lethal dose (LD50) in mice. Epidemiological data suggest that infections by STEC strains that produce only Stx2a progress more often to a life-threatening sequela of infection called hemolytic-uremic syndrome (HUS) than isolates that make Stx1a only or produce both Stx1a and Stx2a. In this study, we found that an E. coli O26:H11 strain that produces both Stx1a and Stx2a was virulent in streptomycin- and ciprofloxacin-treated mice and that mice were protected by administration of an anti-Stx2 antibody. However, we discovered that in the absence of ciprofloxacin, neutralization of Stx1a enhanced the virulence of the strain, a result that corroborated our previous finding that Stx1a reduces the toxicity of Stx2a by the oral route. We further found that intraperitoneal administration of the purified Stx1a B subunit delayed the mean time to death of mice intoxicated with Stx2a and reduced the cytotoxic effect of Stx2a on Vero cells. Taken together, our data suggest that Stx1a reduces both the pathogenicity of Stx2 in vivo and cytotoxicity in vitro.
Collapse
|
41
|
Platnich JM, Muruve DA. NOD-like receptors and inflammasomes: A review of their canonical and non-canonical signaling pathways. Arch Biochem Biophys 2019; 670:4-14. [PMID: 30772258 DOI: 10.1016/j.abb.2019.02.008] [Citation(s) in RCA: 260] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 12/16/2022]
Abstract
The NOD-like receptor (NLR) family of proteins is a group of pattern recognition receptors (PRRs) known to mediate the initial innate immune response to cellular injury and stress. The NLRP proteins represent a fourteen-member subset of the NLR family that contains an N-terminal pyrin domain. Some NLRs are known to form multi-protein complexes known as inflammasomes. Inflammasomes consist of an NLR, the adaptor protein ASC, and the effector molecule pro-caspase-1. Once activated, these inflammasomes facilitate the cleavage and activation of caspase-1, which in turn mediates the cleavage of the pro-inflammatory cytokines IL-1β and IL-18 into their active and secreted forms. Activated caspase-1 also drives the cleavage of gasdermin D, which triggers an inflammatory form of cell death known as pyroptosis. Several NLRs are also known to possess non-canonical, inflammasome-independent functions, regulating a variety of signaling pathways. In this review, a thorough overview of both inflammasome-dependent and -independent NLR signaling will be presented, with highlights from the field as well as promising future directions and postulates based on the known science.
Collapse
Affiliation(s)
- Jaye M Platnich
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Daniel A Muruve
- Department of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
42
|
Sahin E, Bektur E, Baycu C, Burukoglu Dönmez D, Kaygısız B. HYPOTHYROIDISM INCREASES EXPRESSION OF STERILE INFLAMMATION PROTEINS IN RAT HEART TISSUE. ACTA ENDOCRINOLOGICA-BUCHAREST 2019; -5:39-45. [PMID: 31149058 DOI: 10.4183/aeb.2019.39] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Purpose In this study, we aimed to investigate the relationship between hypothyroidism and sterile inflammation in rat heart tissue. Methods Groups; control group (fed with standard rat chow diet and tab water) and the hypothyroid group (fed with a standard rat chow diet and tap water containing 0.05% 6-n-propyl-2-thiouracil for 6-weeks). At the end of the experiment, histopathologic examination was performed. The T3, T4, TSH and myocardial malondialdehyde (MDA) measurements were performed with an ELISA kit. TUNEL assay was performed to demonstrate apoptosis. Sterile inflammation markers, caspase-1 and NLRP3, were investigated by immunohistochemistry and western blot. Results In histopathological examination, we observed leukocyte infiltration, myocardial atrophy, pyknotic nucleated cells and cytoplasmic vacuolization in hypothyroid group whereas the control group showed normal structure. MDA levels in myocardial tissue were significantly high in hypothyroid group when compared to the control group (P<0.05). Myocardial apoptosis increased in hypothyroid group when compared to the control group. NLRP3 and caspase-1 immunoreactivity was higher in the hypothyroid group. In ELISA results, we found significantly higher level of TSH and lower levels of T3 and T4 in hypothyroid group when compared to the control group. Conclusion Hypothyroidism increased oxidative stress, and caused inflammatory alterations in cardiac tissue. In addition, our study also suggested that thyroid hormone deficiency would increase the amounts of cardiac NLRP3 and caspase-1 protein, which indicates that hypothyroidism exerts its destructive effects through sterile inflammation. Elucidation of sterile inflammation-associated pathways may produce promising results in the treatment of hypothyroidism-induced cardiac damage.
Collapse
Affiliation(s)
- E Sahin
- Eskisehir Osmangazi University, Medicine School, Department of Histology and Embryology, Eskisehir, Turkey
| | - E Bektur
- Eskisehir Osmangazi University, Medicine School, Department of Histology and Embryology, Eskisehir, Turkey
| | - C Baycu
- Okan University, Medicine School, Department of Histology and Embryology, Istanbul, Turkey
| | - D Burukoglu Dönmez
- Eskisehir Osmangazi University, Medicine School, Department of Histology and Embryology, Eskisehir, Turkey
| | - B Kaygısız
- Eskisehir Osmangazi University, Medicine School, Department of Pharmacology, Eskisehir, Turkey
| |
Collapse
|
43
|
Cytosolic Recognition of Microbes and Pathogens: Inflammasomes in Action. Microbiol Mol Biol Rev 2018; 82:82/4/e00015-18. [PMID: 30209070 DOI: 10.1128/mmbr.00015-18] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Infection is a dynamic biological process underpinned by a complex interplay between the pathogen and the host. Microbes from all domains of life, including bacteria, viruses, fungi, and protozoan parasites, have the capacity to cause infection. Infection is sensed by the host, which often leads to activation of the inflammasome, a cytosolic macromolecular signaling platform that mediates the release of the proinflammatory cytokines interleukin-1β (IL-1β) and IL-18 and cleavage of the pore-forming protein gasdermin D, leading to pyroptosis. Host-mediated sensing of the infection occurs when pathogens inject or carry pathogen-associated molecular patterns (PAMPs) into the cytoplasm or induce damage that causes cytosolic liberation of danger-associated molecular patterns (DAMPs) in the host cell. Recognition of PAMPs and DAMPs by inflammasome sensors, including NLRP1, NLRP3, NLRC4, NAIP, AIM2, and Pyrin, initiates a cascade of events that culminate in inflammation and cell death. However, pathogens can deploy virulence factors capable of minimizing or evading host detection. This review presents a comprehensive overview of the mechanisms of microbe-induced activation of the inflammasome and the functional consequences of inflammasome activation in infectious diseases. We also explore the microbial strategies used in the evasion of inflammasome sensing at the host-microbe interaction interface.
Collapse
|
44
|
Hughes AJ, Knoten CA, Morris AR, Hauser AR. ASC acts in a caspase-1-independent manner to worsen acute pneumonia caused by Pseudomonas aeruginosa. J Med Microbiol 2018; 67:1168-1180. [PMID: 29957172 DOI: 10.1099/jmm.0.000782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Pseudomonas aeruginosa expresses a type III secretion system (T3SS) that activates the host inflammasome-mediated immune response. We examined the role of inflammasome activation in severe infection outcomes. METHODS We infected C57BL/6 (B6) mice lacking inflammasome components ASC or caspase-1/11 with a highly virulent strain of P. aeruginosa, PSE9, using a mouse model of pneumonia. We evaluated inflammasome activation in vitro by infecting bone marrow-derived macrophages (BMDMs) with PSE9 and measuring cell death and release of inflammasome-dependent cytokines IL-18 and IL-1β. A bioluminescent reporter assay was used to detect activity of caspase-1 and caspase-3/7 in BMDMs from B6 and ASC-deficient mice.Results/Key Findings. ASC-/- mice exhibited significantly improved survival relative to caspase-1/11-/- mice and B6 mice, demonstrating that ASC and caspase-1/11 play differential roles in P. aeruginosa infection. We found that ASC-/- BMDMs exhibited significantly reduced cell death relative to B6 BMDMs, while caspase-1/11-/- BMDMs were resistant to cell death. IL-18 and IL-1β were both detected from supernatants of infected B6 BMDMs, but cytokine release was abrogated in both ASC-/- and caspase-1/11-/- BMDMs. We detected a 2.5-fold increase in the activation of caspase-3/7 in PSE9-infected B6 BMDMs, but no increase in infected ASC-/- BMDMs. Cell death, cytokine release and caspase-3/7 activity were dependent on a functional T3SS. CONCLUSIONS Collectively, these results are consistent with a model whereby the T3SS apparatus of P. aeruginosa activates the caspase-1-dependent inflammasome and caspase-3/7 through an ASC-dependent mechanism. This activation may have implications for the outcomes of P. aeruginosa infections.
Collapse
Affiliation(s)
- A J Hughes
- 1Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C A Knoten
- 1Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,†Present address: Academy for Quality and Safety Improvement, Northwestern Medicine, Chicago, IL, USA
| | - A R Morris
- 1Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - A R Hauser
- 2Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,1Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
45
|
Zhao H, Chen Y, Feng H. P2X7 Receptor-Associated Programmed Cell Death in the Pathophysiology of Hemorrhagic Stroke. Curr Neuropharmacol 2018; 16:1282-1295. [PMID: 29766811 PMCID: PMC6251042 DOI: 10.2174/1570159x16666180516094500] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/17/2017] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
Hemorrhagic stroke is a life-threatening disease characterized by a sudden rupture of cerebral blood vessels, and cell death is widely believed to occur after exposure to blood metabolites or subsequently damaged cells. Recently, programmed cell death, such as apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis, has been demonstrated to play crucial roles in the pathophysiology of stroke. However, the detailed mechanisms of these novel kinds of cell death are still unclear. The P2X7 receptor, previously known for its cytotoxic activity, is an ATP-gated, nonselective cation channel that belongs to the family of ionotropic P2X receptors. Evolving evidence indicates that the P2X7 receptor plays a pivotal role in central nervous system pathology; genetic deletion and pharmacological blockade of the P2X7 receptor provide neuroprotection in various neurological disorders, including intracerebral hemorrhage and subarachnoid hemorrhage. The P2X7 receptor may regulate programmed cell death via (I) exocytosis of secretory lysosomes, (II) exocytosis of autophagosomes or autophagolysosomes during formation of the initial autophagic isolation membrane or omegasome, and (III) direct release of cytosolic IL-1β secondary to regulated cell death by pyroptosis or necroptosis. In this review, we present an overview of P2X7 receptor- associated programmed cell death for further understanding of hemorrhagic stroke pathophysiology, as well as potential therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Hengli Zhao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| |
Collapse
|
46
|
Park JY, Jeong YJ, Park SK, Yoon SJ, Choi S, Jeong DG, Chung SW, Lee BJ, Kim JH, Tesh VL, Lee MS, Park YJ. Shiga Toxins Induce Apoptosis and ER Stress in Human Retinal Pigment Epithelial Cells. Toxins (Basel) 2017; 9:toxins9100319. [PMID: 29027919 PMCID: PMC5666366 DOI: 10.3390/toxins9100319] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 01/03/2023] Open
Abstract
Shiga toxins (Stxs) produced by Shiga toxin-producing bacteria Shigella dysenteriae serotype 1 and select serotypes of Escherichia coli are the most potent known virulence factors in the pathogenesis of hemorrhagic colitis progressing to potentially fatal systemic complications such as acute renal failure, blindness and neurological abnormalities. Although numerous studies have defined apoptotic responses to Shiga toxin type 1 (Stx1) or Shiga toxin type 2 (Stx2) in a variety of cell types, the potential significance of Stx-induced apoptosis of photoreceptor and pigmented cells of the eye following intoxication is unknown. We explored the use of immortalized human retinal pigment epithelial (RPE) cells as an in vitro model of Stx-induced retinal damage. To the best of our knowledge, this study is the first report that intoxication of RPE cells with Stxs activates both apoptotic cell death signaling and the endoplasmic reticulum (ER) stress response. Using live-cell imaging analysis, fluorescently labeled Stx1 or Stx2 were internalized and routed to the RPE cell endoplasmic reticulum. RPE cells were significantly sensitive to wild type Stxs by 72 h, while the cells survived challenge with enzymatically deficient mutant toxins (Stx1A− or Stx2A−). Upon exposure to purified Stxs, RPE cells showed activation of a caspase-dependent apoptotic program involving a reduction of mitochondrial transmembrane potential (Δψm), increased activation of ER stress sensors IRE1, PERK and ATF6, and overexpression CHOP and DR5. Finally, we demonstrated that treatment of RPE cells with Stxs resulted in the activation of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38MAPK), suggesting that the ribotoxic stress response may be triggered. Collectively, these data support the involvement of Stx-induced apoptosis in ocular complications of intoxication. The evaluation of apoptotic responses to Stxs by cells isolated from multiple organs may reveal unique functional patterns of the cytotoxic actions of these toxins in the systemic complications that follow ingestion of toxin-producing bacteria.
Collapse
Affiliation(s)
- Jun-Young Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea.
| | - Yu-Jin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biochemistry, College of Medicine, Konyang University, 158 Gwanjeo-ro, Daejeon 35365, South Korea.
| | - Sung-Kyun Park
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Sung-Jin Yoon
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Song Choi
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Dae Gwin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
| | - Su Wol Chung
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Ulsan 44610, South Korea.
| | - Byung Joo Lee
- Fight Against Angiogenesis-Related Blindness Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea.
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea.
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| | - Moo-Seung Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea.
| | - Young-Jun Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, South Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea.
| |
Collapse
|
47
|
Hall G, Kurosawa S, Stearns-Kurosawa DJ. Shiga Toxin Therapeutics: Beyond Neutralization. Toxins (Basel) 2017; 9:toxins9090291. [PMID: 28925976 PMCID: PMC5618224 DOI: 10.3390/toxins9090291] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 01/04/2023] Open
Abstract
Ribotoxic Shiga toxins are the primary cause of hemolytic uremic syndrome (HUS) in patients infected with Shiga toxin-producing enterohemorrhagic Escherichia coli (STEC), a pathogen class responsible for epidemic outbreaks of gastrointestinal disease around the globe. HUS is a leading cause of pediatric renal failure in otherwise healthy children, resulting in a mortality rate of 10% and a chronic morbidity rate near 25%. There are currently no available therapeutics to prevent or treat HUS in STEC patients despite decades of work elucidating the mechanisms of Shiga toxicity in sensitive cells. The preclinical development of toxin-targeted HUS therapies has been hindered by the sporadic, geographically dispersed nature of STEC outbreaks with HUS cases and the limited financial incentive for the commercial development of therapies for an acute disease with an inconsistent patient population. The following review considers potential therapeutic targeting of the downstream cellular impacts of Shiga toxicity, which include the unfolded protein response (UPR) and the ribotoxic stress response (RSR). Outcomes of the UPR and RSR are relevant to other diseases with large global incidence and prevalence rates, thus reducing barriers to the development of commercial drugs that could improve STEC and HUS patient outcomes.
Collapse
Affiliation(s)
- Gregory Hall
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Shinichiro Kurosawa
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Deborah J Stearns-Kurosawa
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
48
|
Role of NLRP3 Inflammasome in Eosinophilic and Non-eosinophilic Chronic Rhinosinusitis with Nasal Polyps. Inflammation 2017; 39:2045-2052. [PMID: 27614764 DOI: 10.1007/s10753-016-0442-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pathophysiologic mechanisms of human chronic rhinosinusitis with nasal polyps (CRSwNP) remain unclear. We aimed to elucidate expression and biologic role of NLRP3 inflammasome in CRSwNP. Immunohistochemistry (IHC) was conducted to assess NLRP3 immunolabeling, real-time polymerase chain reaction (PCR) was used for IL-9 and NLRP3, and caspase-1 level quantitation in CRSwNP and control subjects. In addition, enzyme-linked immunosorbent assay (ELISA) was employed for analyzing concentrations of IL-1β and IL-18 in the homogenates prepared from tissue specimens. Moreover, human nasal epithelial cells (HNECs) were used to evaluate the effects of lipopolysaccharide (LPS) and glyburide on NLRP3 inflammasome signaling pathway. Results showed that NLRP3 and caspase-1 were overexpressed in CRSwNP, especially in eosinophilic CRSwNP (ECRSwNP). Interestingly, NLRP3 expression had close correlation to that of caspase-1. Concentrations of IL-1β and IL-18 were elevated. NLRP3 inflammasome signaling pathway was augmented by LPS but suppressed by glyburide. In conclusion, NLRP3 inflammasome signaling pathway played a pro-inflammatory role in the pathogenesis of CRSwNP, especially in ECRSwNP. NLRP3 inflammasome signaling pathway was augmented by LPS, but suppressed by glyburide.
Collapse
|
49
|
Ambite I, Puthia M, Nagy K, Cafaro C, Nadeem A, Butler DSC, Rydström G, Filenko NA, Wullt B, Miethke T, Svanborg C. Molecular Basis of Acute Cystitis Reveals Susceptibility Genes and Immunotherapeutic Targets. PLoS Pathog 2016; 12:e1005848. [PMID: 27732661 PMCID: PMC5061333 DOI: 10.1371/journal.ppat.1005848] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/06/2016] [Indexed: 12/19/2022] Open
Abstract
Tissue damage is usually regarded as a necessary price to pay for successful elimination of pathogens by the innate immune defense. Yet, it is possible to distinguish protective from destructive effects of innate immune activation and selectively attenuate molecular nodes that create pathology. Here, we identify acute cystitis as an Interleukin-1 beta (IL-1β)-driven, hyper-inflammatory condition of the infected urinary bladder and IL-1 receptor blockade as a novel therapeutic strategy. Disease severity was controlled by the mechanism of IL-1β processing and mice with intact inflammasome function developed a moderate, self-limiting form of cystitis. The most severe form of acute cystitis was detected in mice lacking the inflammasome constituents ASC or NLRP-3. IL-1β processing was hyperactive in these mice, due to a new, non-canonical mechanism involving the matrix metalloproteinase 7- (MMP-7). ASC and NLRP-3 served as transcriptional repressors of MMP7 and as a result, Mmp7 was markedly overexpressed in the bladder epithelium of Asc-/- and Nlrp3-/- mice. The resulting IL-1β hyper-activation loop included a large number of IL-1β-dependent pro-inflammatory genes and the IL-1 receptor antagonist Anakinra inhibited their expression and rescued susceptible Asc-/- mice from bladder pathology. An MMP inhibitor had a similar therapeutic effect. Finally, elevated levels of IL-1β and MMP-7 were detected in patients with acute cystitis, suggesting a potential role as biomarkers and immunotherapeutic targets. The results reproduce important aspects of human acute cystitis in the murine model and provide a comprehensive molecular framework for the pathogenesis and immunotherapy of acute cystitis, one of the most common infections in man. Infections continue to threaten human health as pathogenic organisms outsmart available therapies with remarkable genetic versatility. Fortunately, microbial versatility is matched by the flexibility of the host immune system which provide a rich source of novel therapeutic concepts. Emerging therapeutic solutions include substances that strengthen the immune system rather than killing the bacteria directly. Selectivity is a concern, however, as boosting of the antibacterial immune response may cause collateral tissue damage. This study addresses how the host response to urinary bladder infection causes acute cystitis and how this response can be attenuated in patients who suffer from this very common condition. We identify the cytokine Interleukin-1 beta (IL-1β) as a key immune response determinant in acute cystitis and successfully treat mice with severe acute cystitis by inhibiting IL-1β or the enzyme MMP-7 that processes IL-1β to its active form. Finally, we detect elevated levels of these molecules in urine samples from patients with cystitis, suggesting clinical relevance and a potential role of IL-1β and MMP-7 both as therapeutic targets and as biomarkers of infection. These findings provide a much needed, molecular framework for the pathogenesis and treatment of acute cystitis.
Collapse
Affiliation(s)
- Ines Ambite
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Manoj Puthia
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Karoly Nagy
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Caterina Cafaro
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Aftab Nadeem
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Daniel S. C. Butler
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Gustav Rydström
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Nina A. Filenko
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Björn Wullt
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thomas Miethke
- Institute of Medical Microbiology and Hygiene, Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Catharina Svanborg
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
50
|
Ogunade I, Arriola K, Jiang Y, Driver J, Staples C, Adesogan A. Effects of 3 sequestering agents on milk aflatoxin M1 concentration and the performance and immune status of dairy cows fed diets artificially contaminated with aflatoxin B1. J Dairy Sci 2016; 99:6263-6273. [DOI: 10.3168/jds.2016-10905] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/12/2016] [Indexed: 01/30/2023]
|