1
|
Firestone K, Gopalakrishna KP, Rogers LM, Peters A, Gaddy JA, Nichols C, Hall MH, Varela HN, Carlin SM, Hillebrand GH, Giacobe EJ, Aronoff DM, Hooven TA. A CRISPRi Library Screen in Group B Streptococcus Identifies Surface Immunogenic Protein (Sip) as a Mediator of Multiple Host Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627252. [PMID: 39677656 PMCID: PMC11643019 DOI: 10.1101/2024.12.06.627252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Group B Streptococcus (GBS; Streptococcus agalactiae) is an important pathobiont capable of colonizing various host environments, contributing to severe perinatal infections. Surface proteins play critical roles in GBS-host interactions, yet comprehensive studies of these proteins' functions have been limited by genetic manipulation challenges. This study leveraged a CRISPR interference (CRISPRi) library to target genes encoding surface-trafficked proteins in GBS, identifying their roles in modulating macrophage cytokine responses. Bioinformatic analysis of 654 GBS genomes revealed 66 conserved surface protein genes. Using a GBS strain expressing chromosomally integrated dCas9, we generated and validated CRISPRi strains targeting these genes. THP-1 macrophage-like cells were exposed to ethanol-killed GBS variants, and pro-inflammatory cytokines TNF-α and IL-1β were measured. Notably, knockdown of the sip gene, encoding the Surface Immunogenic Protein (Sip), significantly increased IL-1β secretion, implicating Sip in caspase-1-dependent regulation. Further, Δsip mutants demonstrated impaired biofilm formation, reduced adherence to human fetal membranes, and diminished uterine persistence in a mouse colonization model. These findings suggest Sip modulates GBS-host interactions critical for pathogenesis, underscoring its potential as a therapeutic target or vaccine component.
Collapse
Affiliation(s)
- K Firestone
- Indiana University School of Medicine, Department of Medicine
| | - K P Gopalakrishna
- California Institute of Technology, Division of Chemistry and Chemical Engineering, Pasadena, CA, USA
| | - L M Rogers
- Indiana University School of Medicine, Department of Medicine
| | - A Peters
- University of Pittsburgh, Dietrich School of Arts and Sciences, Pittsburgh, PA, USA
| | - J A Gaddy
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
- Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, Nashville, TN, USA
- Vanderbilt University, Center for Medicine, Health, and Society, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, TN, USA
| | - C Nichols
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
| | - M H Hall
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, TN, USA
| | - H N Varela
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - S M Carlin
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - G H Hillebrand
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - E J Giacobe
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - D M Aronoff
- Indiana University School of Medicine, Department of Medicine
| | - T A Hooven
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- R.K. Mellon Institute for Pediatric Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Mabry CJ, Weindel CG, Stranahan LW, VanPortfliet JJ, Davis JR, Martinez EL, West AP, Patrick KL, Watson RO. Necrosis drives susceptibility to Mycobacterium tuberculosis in Polg D257A mutator mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603991. [PMID: 39091776 PMCID: PMC11291070 DOI: 10.1101/2024.07.17.603991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The genetic and molecular determinants that underlie the heterogeneity of Mycobacterium tuberculosis (Mtb) infection outcomes in humans are poorly understood. Multiple lines of evidence demonstrate that mitochondrial dysfunction can exacerbate mycobacterial disease severity and mutations in some mitochondrial genes confer susceptibility to mycobacterial infection in humans. Here, we report that mutations in mitochondria DNA (mtDNA) polymerase gamma (POLG) potentiate susceptibility to Mtb infection in mice. PolgD257A mutator mtDNA mice fail to mount a protective innate immune response at an early infection timepoint, evidenced by high bacterial burdens, reduced M1 macrophages, and excessive neutrophil infiltration in the lungs. Immunohistochemistry reveals signs of enhanced necrosis in the lungs of Mtb-infected PolgD257A mice and PolgD257A mutator macrophages are hyper-susceptible to extrinsic triggers of necroptosis ex vivo. By assigning a role for mtDNA mutations in driving necrosis during Mtb infection, this work further highlights the requirement for mitochondrial homeostasis in mounting balanced immune responses to Mtb.
Collapse
Affiliation(s)
- CJ Mabry
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - CG Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - LW Stranahan
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA
| | - JJ VanPortfliet
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - JR Davis
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - EL Martinez
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
| | - AP West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - KL Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - RO Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
3
|
Mohammed WH, Sulaiman GM, Abomughaid MM, Klionsky DJ, Abu-Alghayth MH. The dual role of autophagy in suppressing and promoting hepatocellular carcinoma. Front Cell Dev Biol 2024; 12:1472574. [PMID: 39463763 PMCID: PMC11502961 DOI: 10.3389/fcell.2024.1472574] [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: 08/01/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
The 5-year survival rate for hepatocellular carcinoma (HCC), a deadly form of liver cancer, is quite low. Although drug therapy is successful, patients with advanced liver cancer frequently develop resistance because of the significant phenotypic and genetic heterogeneity of these cells. The overexpression of drug efflux transporters, downstream adaptive responses, malfunctioning DNA damage repair, epigenetic modification, the tumor microenvironment, and the extracellular matrix can all be linked to drug resistance. The evolutionary process of autophagy, which is in charge of intracellular breakdown, is intimately linked to medication resistance in HCC. Autophagy is involved in both the promotion and suppression of cancer by influencing treatment resistance, metastasis, carcinogenesis, and the viability of stem cells. Certain autophagy regulators are employed in anticancer treatment; however, because of the dual functions of autophagy, their use is restricted, and therapeutic failure is increased. By focusing on autophagy, it is possible to reduce HCC expansion and metastasis, and enhance tumor cell reactivity to treatment. Macroautophagy, the best-characterized type of autophagy, involves the formation of a sequestering compartment termed a phagophore, which surrounds and encloses aberrant or superfluous components. The phagophore matures into a double-membrane autophagosome that delivers the cargo to the lysosome; lysosomes and autophagosomes fuse to degrade and recycle the cargo. Macroautophagy plays dual functions in both promoting and suppressing cancer in a variety of cancer types.
Collapse
Affiliation(s)
- Wasnaa H. Mohammed
- Department of Biotechnology, College of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Ghassan M. Sulaiman
- Department of Biotechnology, College of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Mosleh M. Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Mohammed H. Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
4
|
Tan X, Long Y, Zhang R, Zhang Y, You Z, Yang L. Punicalagin Ameliorates Diabetic Liver Injury by Inhibiting Pyroptosis and Promoting Autophagy via Modulation of the FoxO1/TXNIP Signaling Pathway. Mol Nutr Food Res 2024; 68:e2300912. [PMID: 38847553 DOI: 10.1002/mnfr.202300912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/29/2024] [Indexed: 07/04/2024]
Abstract
Diabetic liver injury (DLI) is one of the complications of diabetes mellitus, which seriously jeopardizes human health. Punicalagin (PU), a polyphenolic compound mainly found in pomegranate peel, has been shown to ameliorate metabolic diseases such as DLI, and the mechanism needs to be further explored. In this study, a HFD/STZ-induced diabetic mouse model is established to investigate the effect and mechanism of PU on DLI. The results show that PU intervention significantly improves liver histology and serum biochemical abnormalities in diabetic mice, significantly inhibits the expression of pyroptosis-related proteins such as NLRP3, Caspase1, IL-1β, and GSDMD in the liver of diabetic mice, and up-regulated the expression of autophagy-related proteins. Meanwhile, PU treatment significantly increases FoxO1 protein expression and inhibits TXNIP protein expression in the liver of diabetic mice. The above results are further verified in the HepG2 cell injury model induced by high glucose. AS1842856 is a FoxO1 specific inhibitor. The intervention of AS1842856 combined with PU reverses the regulatory effects of PU on pyroptosis and autophagy in HepG2 cells. In conclusion, this study demonstrates that PU may inhibit pyroptosis and upregulate autophagy by regulating FoxO1/TXNIP signaling, thereby alleviating DLI.
Collapse
Affiliation(s)
- Xiuying Tan
- Xiangya School of Public Health, Central South University, Changsha, 410013, China
| | - Yi Long
- Children's Medical Center, People's Hospital, Hunan Province, Changsha, 410005, China
| | - Rou Zhang
- Xiangya School of Public Health, Central South University, Changsha, 410013, China
| | - Yuhan Zhang
- Xiangya School of Public Health, Central South University, Changsha, 410013, China
| | - Ziyi You
- Xiangya School of Public Health, Central South University, Changsha, 410013, China
| | - Lina Yang
- Xiangya School of Public Health, Central South University, Changsha, 410013, China
| |
Collapse
|
5
|
Wang Y, Chen G, Xu M, Cui Y, He W, Zeng H, Zeng T, Cheng R, Li X. Caspase-1 Deficiency Modulates Adipogenesis through Atg7-Mediated Autophagy: An Inflammatory-Independent Mechanism. Biomolecules 2024; 14:501. [PMID: 38672517 PMCID: PMC11048440 DOI: 10.3390/biom14040501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity stands as a significant risk factor for type 2 diabetes, hyperlipidemia, and cardiovascular diseases, intertwining increased inflammation and decreased adipogenesis with metabolic disorders. Studies have highlighted the correlation between Caspase-1 and inflammation in obesity, elucidating its essential role in the biological functions of adipose tissue. However, the impact of Caspase-1 on adipogenesis and the underlying mechanisms remain largely elusive. In our study, we observed a positive correlation between Caspase-1 expression and obesity and its association with adipogenesis. In vivo experiments revealed that, under normal diet conditions, Caspase-1 deficiency improved glucose homeostasis, stimulated subcutaneous adipose tissue expansion, and enhanced adipogenesis. Furthermore, our findings indicate that Caspase-1 deficiency promotes the expression of autophagy-related proteins and inhibits autophagy with 3-MA or CQ blocked Caspase-1 deficiency-induced adipogenesis in vitro. Notably, Caspase-1 deficiency promotes adipogenesis via Atg7-mediated autophagy activation. In addition, Caspase-1 deficiency resisted against high-fat diet-induced obesity and glucose intolerance. Our study proposes the downregulation of Caspase-1 as a promising strategy for mitigating obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rui Cheng
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
6
|
Bullen CK, Singh AK, Krug S, Lun S, Thakur P, Srikrishna G, Bishai WR. MDA5 RNA-sensing pathway activation by Mycobacterium tuberculosis promotes innate immune subversion and pathogen survival. JCI Insight 2023; 8:e166242. [PMID: 37725440 PMCID: PMC10619499 DOI: 10.1172/jci.insight.166242] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 09/13/2023] [Indexed: 09/21/2023] Open
Abstract
Host cytosolic sensing of Mycobacterium tuberculosis (M. tuberculosis) RNA by the RIG-I-like receptor (RLR) family perturbs innate immune control within macrophages; however, a distinct role of MDA5, a member of the RLR family, in M. tuberculosis pathogenesis has yet to be fully elucidated. To further define the role of MDA5 in M. tuberculosis pathogenesis, we evaluated M. tuberculosis intracellular growth and innate immune responses in WT and Mda5-/- macrophages. Transfection of M. tuberculosis RNA strongly induced proinflammatory cytokine production in WT macrophages, which was abrogated in Mda5-/- macrophages. M. tuberculosis infection in macrophages induced MDA5 protein expression, accompanied by an increase in MDA5 activation as assessed by multimer formation. IFN-γ-primed Mda5-/- macrophages effectively contained intracellular M. tuberculosis proliferation to a markedly greater degree than WT macrophages. Further comparisons of WT versus Mda5-/- macrophages revealed that during M. tuberculosis infection MDA5 contributed to IL-1β production and inflammasome activation and that loss of MDA5 led to a substantial increase in autophagy. In the mouse TB model, loss of MDA5 conferred host survival benefits with a concomitant reduction in M. tuberculosis bacillary burden. These data reveal that loss of MDA5 is host protective during M. tuberculosis infection in vitro and in vivo, suggesting that M. tuberculosis exploits MDA5 to subvert immune containment.
Collapse
|
7
|
Kanika, Khan R. Functionalized nanomaterials targeting NLRP3 inflammasome driven immunomodulation: Friend or Foe. NANOSCALE 2023; 15:15906-15928. [PMID: 37750698 DOI: 10.1039/d3nr03857b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The advancement in drug delivery systems in recent times has significantly enhanced therapeutic effects by enabling site-specific targeting through nanocarriers. These nanocarriers serve as invaluable tools for pharmacotherapeutic advancements against various disorders that enhance the effectiveness of encapsulated drugs by reducing their toxicity and increasing the efficacy of less potent drugs, thereby improving the therapeutic index. Inflammasomes, protein complexes located in the activated immune cell cytoplasm, regulate the activation of caspases involved in inflammation. However, aberrant activation of inflammasomes can result in uncontrolled tissue responses, contributing to the development of various diseases. Therefore, achieving a precise balance between inflammasome inhibition and activation is crucial for effectively treating inflammatory disorders through targeted functionalized nanocarriers. Despite the wealth of available data on the relevance of functionalized nanocarriers in inflammatory disorders, the nanotechnological potential to modulate inflammasomes has not been adequately explored. In this comprehensive review, we highlight the latest research on the modulation of the inflammasome cascade, both upregulating and downregulating its function, using nanocarriers in the context of inflammatory disorders. The utilization of nanocarriers as a therapeutic strategy holds immense potential for researchers aiming to effectively target and modulate inflammasomes in the treatment of inflammatory disorders, thus improving disease severity outcomes.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| |
Collapse
|
8
|
Yue L, Cao H, Qi J, Yuan J, Wang X, Wang Y, Shan B, Ke H, Li H, Luan N, Liu C. Pretreatment with 3-methyladenine ameliorated Pseudomonas aeruginosa-induced acute pneumonia by inhibiting cell death of neutrophils in a mouse infection model. Int J Med Microbiol 2023; 313:151574. [PMID: 36736016 DOI: 10.1016/j.ijmm.2023.151574] [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: 02/07/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/21/2023] Open
Abstract
Pseudomonas aeruginosa is one of the leading causes of nosocomial infections worldwide. Clinical isolates that are resistant to multiple antimicrobials make it intractable. The interactions between P. aeruginosa and host cell death have multiple effects on bacterial clearance and inflammation; however, the potential intervention effects remain to be defined. Herein, we demonstrated that intravenous administration of 3-methyladenine before, but not after, P. aeruginosa infection enhanced autophagy-independent survival, which was accompanied by a decrease in the bacterial load, alleviation of pathology and reduction in inflammatory cytokines, in an acute pneumonia mouse model. Interestingly, these beneficial effects were not dependent on neutrophil recruitment or phagocytosis, but on the enhanced killing capacity induced by inhibiting the cell death of 3-MA pretreated neutrophils. These findings demonstrate a novel protective role of 3-MA pretreatment in P. aeruginosa-induced acute pneumonia.
Collapse
Affiliation(s)
- Lei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Jialong Qi
- The First People's Hospital of Yunnan Province & Affiliated Hospital of Kunming University of Science and Technology, Kunming 650034, China
| | - Jin Yuan
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Xin Wang
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Yunfei Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Bin Shan
- Department of Clinical Lab, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Huaxin Ke
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Hua Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Ning Luan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| |
Collapse
|
9
|
Panda C, Mahapatra RK. Bi-Directional Relationship Between Autophagy and Inflammasomes in Neurodegenerative Disorders. Cell Mol Neurobiol 2023; 43:115-137. [PMID: 35066716 DOI: 10.1007/s10571-021-01184-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/12/2021] [Indexed: 01/18/2023]
Abstract
The innate immune system, as the first line of cellular defense, triggers a protective response called inflammation when encountered with invading pathogens. Inflammasome is a multi-protein cytosolic signaling complex that induces inflammation and is critical for inflammation-induced pyroptotic cell death. Inflammasome activation has been found associated with neurodegenerative disorders (NDs), inflammatory diseases, and cancer. Autophagy is a crucial intracellular quality control and homeostasis process which removes the dysfunctional organelles, damaged proteins, and pathogens by sequestering the cytosolic components in a double-membrane vesicle, which eventually fuses with lysosome resulting in cargo degradation. Autophagy disruption has been observed in many NDs presented with persistent neuroinflammation and excessive inflammasome activation. An interplay between inflammation activation and the autophagy process has been realized over the last decade. In the case of NDs, autophagy regulates neuroinflammation load and cellular damage either by engulfing the misfolded protein deposits, dysfunctional mitochondria, or the inflammasome complex itself. A healthy two-way regulation between both cellular processes has been realized for cell survival and cell defense during inflammatory conditions. Therefore, clinical interest in the modulation of inflammasome activation by autophagy inducers is rapidly growing. In this review, we discuss the structural basis of inflammasome activation and the mechanistic ideas of the autophagy process in NDs. Along with comments on multiple ways of neuroinflammation regulation by microglial autophagy, we also present a perspective on pharmacological opportunities in this molecular interplay pertaining to NDs.
Collapse
Affiliation(s)
- Chinmaya Panda
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India
| | - Rajani Kanta Mahapatra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
10
|
NLRP3 Inflammasome: From Pathophysiology to Therapeutic Target in Major Depressive Disorder. Int J Mol Sci 2022; 24:ijms24010133. [PMID: 36613574 PMCID: PMC9820112 DOI: 10.3390/ijms24010133] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Major depressive disorder (MDD) is a highly prevalent psychiatric disorder, whose pathophysiology has been linked to the neuroinflammatory process. The increased activity of the Nod-like receptor pyrin containing protein 3 (NLRP3) inflammasome, an intracellular multiprotein complex, is intrinsically implicated in neuroinflammation by promoting the maturation and release of proinflammatory cytokines such as interleukin (IL)-1β and IL-18. Interestingly, individuals suffering from MDD have higher expression of NLRP3 inflammasome components and proinflammatory cytokines when compared to healthy individuals. In part, intense activation of the inflammasome may be related to autophagic impairment. Noteworthy, some conventional antidepressants induce autophagy, resulting in less activation of the NLRP3 inflammasome. In addition, the fast-acting antidepressant ketamine, some bioactive compounds and physical exercise have also been shown to have anti-inflammatory properties via inflammasome inhibition. Therefore, it is suggested that modulation of inflammasome-driven pathways may have an antidepressant effect. Here, we review the role of the NLRP3 inflammasome in the pathogenesis of MDD, highlighting that pathways related to its priming and activation are potential therapeutic targets for the treatment of MDD.
Collapse
|
11
|
Yin J, Gong G, Wan W, Liu X. Pyroptosis in spinal cord injury. Front Cell Neurosci 2022; 16:949939. [PMID: 36467606 PMCID: PMC9715394 DOI: 10.3389/fncel.2022.949939] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/03/2022] [Indexed: 10/21/2023] Open
Abstract
Spinal cord injury (SCI) often brings devastating consequences to patients and their families. Pathophysiologically, the primary insult causes irreversible damage to neurons and glial cells and initiates the secondary damage cascade, further leading to inflammation, ischemia, and cells death. In SCI, the release of various inflammatory mediators aggravates nerve injury. Pyroptosis is a new pro-inflammatory pattern of regulated cell death (RCD), mainly mediated by caspase-1 or caspase-11/4/5. Gasdermins family are pore-forming proteins known as the executor of pyroptosis and the gasdermin D (GSDMD) is best characterized. Pyroptosis occurs in multiple central nervous system (CNS) cell types, especially plays a vital role in the development of SCI. We review here the evidence for pyroptosis in SCI, and focus on the pyroptosis of different cells and the crosstalk between them. In addition, we discuss the interaction between pyroptosis and other forms of RCD in SCI. We also summarize the therapeutic strategies for pyroptosis inhibition, so as to provide novel ideas for improving outcomes following SCI.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenhui Wan
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Liang L, Wang H, Hu Y, Bian H, Xiao L, Wang G. Oridonin relieves depressive-like behaviors by inhibiting neuroinflammation and autophagy impairment in rats subjected to chronic unpredictable mild stress. Phytother Res 2022; 36:3335-3351. [PMID: 35686337 DOI: 10.1002/ptr.7518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/21/2022] [Accepted: 05/17/2022] [Indexed: 11/11/2022]
Abstract
Major depressive disorder (MDD) is a severe life-threatening disorder with increasing prevalence. However, the mechanistic interplay between depression, neuroinflammation, and autophagy is yet to be demonstrated. This study investigated the effect of Oridonin on CUMS-induced depression, neuroinflammation, and autophagy impairment. Male 4-week-old Sprague-Dawley rats were subjected to chronic unpredictable mild stress (CUMS), some of which were injected with Oridonin, fluoxetine (FLX), or their combination at different durations of CUMS. CUMS significantly increased the levels of cytokines (IL-1β, IL-18, and caspase-1), reduced autophagy-related protein levels (Beclin-1, p62, Atg5, and LC3B), and caused microglia cells activation. Oridonin prevented and reversed the depressive-like behavior. Furthermore, it has a stronger and longer-lasting antidepressant effect than FLX. And the antidepressant effect of Oridonin in combination with fluoxetine was greater than that of high-dose fluoxetine alone. In addition, Oridonin significantly normalized autophagy-related protein levels, and reduced levels of cytokines by blocking the interaction between NLRP3 and NEK7. Similarly, Oridonin abolished levels of cytokines and reversed autophagy impairment in LPS-activated BV2 cells. All these results supported our hypothesis that Oridonin possesses potent anti-depressive action, which might be mediated via inhibition of neuroinflammation and autophagy impairment by blocking the interaction between NLRP3 and NEK7.
Collapse
Affiliation(s)
- Liang Liang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Psychology, The Fourth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Hui Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Hu
- Department of Psychology, The Fourth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Hetao Bian
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Wang H, Xie Z, Yang F, Wang Y, Jiang H, Huang X, Zhang Y. Salmonella enterica serovar Typhi influences inflammation and autophagy in macrophages. Braz J Microbiol 2022; 53:525-534. [PMID: 35274232 PMCID: PMC9151981 DOI: 10.1007/s42770-022-00719-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) is a human enteropathogen that can survive in macrophages and cause systemic infection. Autophagy and inflammation are two important immune responses of macrophages that contribute to the elimination of pathogens. However, Salmonella has derived many strategies to evade inflammation and autophagy. This study investigated inflammation-related NF-κB signaling pathways and autophagy in S. Typhi-infected macrophages. RNA-seq and quantitative real-time PCR indicated that mRNA levels of NF-κB signaling pathway and autophagy-related genes were dynamically influenced in S. Typhi-infected macrophages. Western blots revealed that S. Typhi activated the NF-κB signaling pathway and induced the expression of inhibitor protein IκBζ. In addition, S. Typhi enhanced autophagy during early stages of infection and may inhibit autophagy during late stages of infection. Thus, we propose that S. Typhi can influence the NF-κB signaling pathway and autophagy in macrophages.
Collapse
Affiliation(s)
- Huiyun Wang
- JiangYin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu, China
| | - Zhongyi Xie
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Fanfan Yang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yurou Wang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Haiqiang Jiang
- JiangYin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu, China
| | - Xinxiang Huang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
14
|
Ribeiro VR, Romao-Veiga M, Nunes PR, Peracoli JC, Peracoli MTS. Increase of autophagy marker p62 in the placenta from pregnant women with preeclampsia. Hum Immunol 2022; 83:447-452. [PMID: 35210117 DOI: 10.1016/j.humimm.2022.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 11/04/2022]
Abstract
Preeclampsia (PE) is a multisystemic disorder characterized by abnormal placentation. Autophagy is a lysosomal degradation pathway that removes protein aggregates and damaged organelles, and it seems to be essential for cell survival during stress, hypoxia, and for implantation and development of the placenta. p62/SQSTM1 is an autophagy marker that not only binds proteins destined for elimination but is also constitutively degraded by this mechanism. Considering that the placenta plays an important role in the pathogenesis of PE, the present study aimed to evaluate the gene and protein expression of p62/SQSTM1 in placentas from pregnant women with PE. Placental tissues from 20 women with PE classified into three groups according to gestational age, 27-31 weeks (n = 8); 32-36 weeks (n = 6); 37-39 weeks (n = 6), and 20 normotensives (NT) pregnant women were collected and employed for p62/SQSTM1 expression by quantitative polymerase chain reaction (qPCR), immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) techniques. p62/SQSTM1 mRNA levels were significantly lower, while protein expression was significantly higher in the placenta of pregnant women with PE than in NT pregnant women, and these results remained similar after separating the groups by gestational age. In conclusion, the results suggest that there is a reduction of autophagic activity in pregnant women with PE. Studies involving cross-talk between autophagy, inflammasomes, nuclear transcription factor (NF-κB) activation pathways, and aggregation of protein in the placenta from women with PE might help to better understand the pathogenesis of this important obstetric pathology.
Collapse
Affiliation(s)
- Vanessa Rocha Ribeiro
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University, Botucatu Sao Paulo, Brazil.
| | - Mariana Romao-Veiga
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University, Botucatu Sao Paulo, Brazil
| | - Priscila Rezeck Nunes
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University, Botucatu Sao Paulo, Brazil
| | - Jose Carlos Peracoli
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University, Botucatu Sao Paulo, Brazil
| | - Maria Terezinha Serrao Peracoli
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University, Botucatu Sao Paulo, Brazil; Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu Sao Paulo, Brazil
| |
Collapse
|
15
|
Chen YS, Chuang WC, Kung HN, Cheng CY, Huang DY, Sekar P, Lin WW. Pan-Caspase Inhibitor zVAD Induces Necroptotic and Autophagic Cell Death in TLR3/4-Stimulated Macrophages. Mol Cells 2022; 45:257-272. [PMID: 34949739 PMCID: PMC9001149 DOI: 10.14348/molcells.2021.0193] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/24/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022] Open
Abstract
In addition to inducing apoptosis, caspase inhibition contributes to necroptosis and/or autophagy depending on the cell type and cellular context. In macrophages, necroptosis can be induced by co-treatment with Toll-like receptor (TLR) ligands (lipopolysaccharide [LPS] for TLR4 and polyinosinic-polycytidylic acid [poly I:C] for TLR3) and a cell-permeable pan-caspase inhibitor zVAD. Here, we elucidated the signaling pathways and molecular mechanisms of cell death. We showed that LPS/zVAD- and poly I:C/zVAD-induced cell death in bone marrow-derived macrophages (BMDMs) was inhibited by receptor-interacting protein kinase 1 (RIP1) inhibitor necrostatin-1 and autophagy inhibitor 3-methyladenine. Electron microscopic images displayed autophagosome/autolysosomes, and immunoblotting data revealed increased LC3II expression. Although zVAD did not affect LPS- or poly I:C-induced activation of IKK, JNK, and p38, it enhanced IRF3 and STAT1 activation as well as type I interferon (IFN) expression. In addition, zVAD inhibited ERK and Akt phosphorylation induced by LPS and poly I:C. Of note, zVAD-induced enhancement of the IRF3/IFN/STAT1 axis was abolished by necrostatin-1, while zVAD-induced inhibition of ERK and Akt was not. Our data further support the involvement of autocrine IFNs action in reactive oxygen species (ROS)-dependent necroptosis, LPS/zVAD-elicited ROS production was inhibited by necrostatin-1, neutralizing antibody of IFN receptor (IFNR) and JAK inhibitor AZD1480. Accordingly, both cell death and ROS production induced by TLR ligands plus zVAD were abrogated in STAT1 knockout macrophages. We conclude that enhanced TRIF-RIP1-dependent autocrine action of IFNβ, rather than inhibition of ERK or Akt, is involved in TLRs/zVAD-induced autophagic and necroptotic cell death via the JAK/STAT1/ROS pathway.
Collapse
Affiliation(s)
- Yuan-Shen Chen
- Department of Neurosurgery, National Taiwan University Hospital Yunlin Branch, Douliu 64041, Taiwan
| | - Wei-Chu Chuang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Hsiu-Ni Kung
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Ching-Yuan Cheng
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Ponarulselvam Sekar
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
16
|
CrkII/Abl phosphorylation cascade is critical for NLRC4 inflammasome activity and is blocked by Pseudomonas aeruginosa ExoT. Nat Commun 2022; 13:1295. [PMID: 35277504 PMCID: PMC8917168 DOI: 10.1038/s41467-022-28967-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/22/2022] [Indexed: 11/17/2022] Open
Abstract
Type 3 Secretion System (T3SS) is a highly conserved virulence structure that plays an essential role in the pathogenesis of many Gram-negative pathogenic bacteria, including Pseudomonas aeruginosa. Exotoxin T (ExoT) is the only T3SS effector protein that is expressed in all T3SS-expressing P. aeruginosa strains. Here we show that T3SS recognition leads to a rapid phosphorylation cascade involving Abl / PKCδ / NLRC4, which results in NLRC4 inflammasome activation, culminating in inflammatory responses that limit P. aeruginosa infection in wounds. We further show that ExoT functions as the main anti-inflammatory agent for P. aeruginosa in that it blocks the phosphorylation cascade through Abl / PKCδ / NLRC4 by targeting CrkII, which we further demonstrate to be important for Abl transactivation and NLRC4 inflammasome activation in response to T3SS and P. aeruginosa infection. Pseudomonas aeruginosa secretes the toxin ExoT, which is important for pathogenesis. Here, the authors show that ExoT inhibits NLRC4-dependent inflammatory responses during wound infection.
Collapse
|
17
|
Avbelj M, Hafner-Bratkovič I, Lainšček D, Manček-Keber M, Peternelj TT, Panter G, Treon SP, Gole B, Potočnik U, Jerala R. Cleavage-Mediated Regulation of Myd88 Signaling by Inflammasome-Activated Caspase-1. Front Immunol 2022; 12:790258. [PMID: 35069570 PMCID: PMC8767097 DOI: 10.3389/fimmu.2021.790258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/15/2021] [Indexed: 01/07/2023] Open
Abstract
Coordination among multiple signaling pathways ensures an appropriate immune response, where a signaling pathway may impair or augment another signaling pathway. Here, we report a negative feedback regulation of signaling through the key innate immune mediator MyD88 by inflammasome-activated caspase-1. NLRP3 inflammasome activation impaired agonist- or infection-induced TLR signaling and cytokine production through the proteolytic cleavage of MyD88 by caspase-1. Site-specific mutagenesis was used to identify caspase-1 cleavage site within MyD88 intermediary segment. Different cleavage site location within MyD88 defined the functional consequences of MyD88 cleavage between mouse and human cells. LPS/monosodium urate–induced mouse inflammation model corroborated the physiological role of this mechanism of regulation, that could be reversed by chemical inhibition of NLRP3. While Toll/interleukin-1 receptor (TIR) domain released by MyD88 cleavage additionally contributed to the inhibition of signaling, Waldenström’s macroglobulinemia associated MyD88L265P mutation is able to evade the caspase-1-mediated inhibition of MyD88 signaling through the ability of its TIRL265P domain to recruit full length MyD88 and facilitate signaling. The characterization of this mechanism reveals an additional layer of innate immunity regulation.
Collapse
Affiliation(s)
- Monika Avbelj
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.,EN-FIST Centre of Excellence, Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.,EN-FIST Centre of Excellence, Ljubljana, Slovenia
| | - Mateja Manček-Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.,EN-FIST Centre of Excellence, Ljubljana, Slovenia
| | - Tina Tinkara Peternelj
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Gabriela Panter
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Steven P Treon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Boris Gole
- Centre for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Uroš Potočnik
- Centre for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Laboratory of Biochemistry, Molecular Biology and Genomics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.,EN-FIST Centre of Excellence, Ljubljana, Slovenia
| |
Collapse
|
18
|
Zhu L, Liu L. New Insights Into the Interplay Among Autophagy, the NLRP3 Inflammasome and Inflammation in Adipose Tissue. Front Endocrinol (Lausanne) 2022; 13:739882. [PMID: 35432210 PMCID: PMC9008752 DOI: 10.3389/fendo.2022.739882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is a feature of metabolic syndrome with chronic inflammation in obese subjects, characterized by adipose tissue (AT) expansion, proinflammatory factor overexpression, and macrophage infiltration. Autophagy modulates inflammation in the enlargement of AT as an essential step for maintaining the balance in energy metabolism and waste elimination. Signaling originating from dysfunctional AT, such as AT containing hypertrophic adipocytes and surrounding macrophages, activates NOD-like receptor family 3 (NLRP3) inflammasome. There are interactions about altered autophagy and NLRP3 inflammasome activation during the progress in obesity. We summarize the current studies and potential mechanisms associated with autophagy and NLRP3 inflammasome in AT inflammation and aim to provide further evidence for research on obesity and obesity-related complications.
Collapse
Affiliation(s)
- Liyuan Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
- *Correspondence: Ling Liu,
| |
Collapse
|
19
|
Chang P, Li H, Hu H, Li Y, Wang T. The Role of HDAC6 in Autophagy and NLRP3 Inflammasome. Front Immunol 2021; 12:763831. [PMID: 34777380 PMCID: PMC8578992 DOI: 10.3389/fimmu.2021.763831] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy fights against harmful stimuli and degrades cytosolic macromolecules, organelles, and intracellular pathogens. Autophagy dysfunction is associated with many diseases, including infectious and inflammatory diseases. Recent studies have identified the critical role of the NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasomes activation in the innate immune system, which mediates the secretion of proinflammatory cytokines IL-1β/IL-18 and cleaves Gasdermin D to induce pyroptosis in response to pathogenic and sterile stimuli. Accumulating evidence has highlighted the crosstalk between autophagy and NLRP3 inflammasome in multifaceted ways to influence host defense and inflammation. However, the underlying mechanisms require further clarification. Histone deacetylase 6 (HDAC6) is a class IIb deacetylase among the 18 mammalian HDACs, which mainly localizes in the cytoplasm. It is involved in two functional deacetylase domains and a ubiquitin-binding zinc finger domain (ZnF-BUZ). Due to its unique structure, HDAC6 regulates various physiological processes, including autophagy and NLRP3 inflammasome, and may play a role in the crosstalk between them. In this review, we provide insight into the mechanisms by which HDAC6 regulates autophagy and NLRP3 inflammasome and we explored the possibility and challenges of HDAC6 in the crosstalk between autophagy and NLRP3 inflammasome. Finally, we discuss HDAC6 inhibitors as a potential therapeutic approach targeting either autophagy or NLRP3 inflammasome as an anti-inflammatory strategy, although further clarification is required regarding their crosstalk.
Collapse
Affiliation(s)
- Panpan Chang
- Trauma Medicine Center, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration (Peking University), National Center for Trauma Medicine of China, Beijing, China
| | - Hao Li
- Department of Emergency, First Hospital of China Medical University, Shenyang, China
| | - Hui Hu
- Department of Traumatology, Central Hospital of Chongqing University, Chongqing Emergency Medical Center, Chongqing, China
| | - Yongqing Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Tianbing Wang
- Trauma Medicine Center, Peking University People's Hospital, Key Laboratory of Trauma and Neural Regeneration (Peking University), National Center for Trauma Medicine of China, Beijing, China
| |
Collapse
|
20
|
Flores-Vega VR, Vargas-Roldán SY, Lezana-Fernández JL, Lascurain R, Santos-Preciado JI, Rosales-Reyes R. Bacterial Subversion of Autophagy in Cystic Fibrosis. Front Cell Infect Microbiol 2021; 11:760922. [PMID: 34692569 PMCID: PMC8531276 DOI: 10.3389/fcimb.2021.760922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease affecting more than 70,000 people worldwide. It is caused by a mutation in the cftr gene, a chloride ion transporter localized in the plasma membrane of lung epithelial cells and other organs. The loss of CFTR function alters chloride, bicarbonate, and water transport through the plasma membrane, promoting the production of a thick and sticky mucus in which bacteria including Pseudomonas aeruginosa and Burkholderia cenocepacia can produce chronic infections that eventually decrease the lung function and increase the risk of mortality. Autophagy is a well-conserved lysosomal degradation pathway that mediates pathogen clearance and plays an important role in the control of bacterial infections. In this mini-review, we describe the principal strategies used by P. aeruginosa and B. cenocepacia to survive and avoid microbicidal mechanisms within the autophagic pathway leading to the establishment of chronic inflammatory immune responses that gradually compromise the lung function and the life of CF patients.
Collapse
Affiliation(s)
- Verónica Roxana Flores-Vega
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Escuela de Ciencias de la Salud, Universidad del Valle de México, Campus Coyoacán, Mexico City, Mexico
| | - Silvia Yalid Vargas-Roldán
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Mexico City, Mexico
| | - José Luis Lezana-Fernández
- Laboratorio de Fisiología Respiratoria y la Clínica de Fibrosis Quística, Hospital Infantil de México Federico Gómez, Mexico City, Mexico.,Dirección Médica, Asociación Mexicana de Fibrosis Quística, Mexico City, Mexico
| | - Ricardo Lascurain
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Ignacio Santos-Preciado
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Roberto Rosales-Reyes
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
21
|
Baicalin alleviates Mycoplasma gallisepticum-induced oxidative stress and inflammation via modulating NLRP3 inflammasome-autophagy pathway. Int Immunopharmacol 2021; 101:108250. [PMID: 34656906 DOI: 10.1016/j.intimp.2021.108250] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 12/16/2022]
Abstract
Baicalin is a well-known flavonoid compound, possess therapeutic potential against inflammatory diseases. Previous studies reported that Mycoplasma gallisepticum (MG) induced inflammatory response and immune dysregulation inside the host body. However, the underlying molecular mechanisms of baicalin against MG-infected chicken-like macrophages (HD11 cells) are still illusive. Oxidant status and total reactive oxygen species (ROS) were detected by ELISA assays and flow cytometry respectively. Mitochondrial membrane potential (ΔΨM) was evaluated by immunofluorescence microscopy. Transmission electron microscopy was used for ultrastructural analysis. The hallmarks of inflammation and autophagy were determined by western blotting. Oxidative stress and reactive oxygen species (ROS) were significantly enhanced in the MG-infected HD11 cells. MG infection caused disruption in the mitochondrial membrane potential (ΔΨM) compared to the control conditions. Meanwhile, baicalin treatment reduced MG-induced reactive oxygen species (ROS), oxidative stress and alleviated the disruption in ΔΨM. The activities of inflammatory markers were significantly enhanced in the MG-infected HD11 cells. Increased protein expressions of TLR-2-NF-κB pathway, NLRP3-inflammasome and autophagy-related proteins were detected in the MG-infected HD11 cells. Besides, baicalin treatment significantly reduced the protein expressions of TLR-2-NF-κB pathway and NLRP3 inflammasome. While, the autophagy-related proteins were significantly enhanced with baicalin treatment in a dose-dependent manner in the MG-infected HD11 cells. The results showed that baicalin prevented HD11 cells from MG-induced oxidative stress and inflammation via the opposite modulation of TLR-2-NF-κB-mediated NLRP3-inflammasome pathway and autophagy, and baicalin could be a promising candidate for the prevention of inflammatory effects caused by MG-infection in macrophages.
Collapse
|
22
|
Jiang L, Luo S, Qiu T, Li Q, Jiang C, Sun X, Yang G, Zhang C, Liu X, Jiang L. Bidirectional role of reactive oxygen species during inflammasome activation in acrolein-induced human EAhy926 cells pyroptosis. Toxicol Mech Methods 2021; 31:680-689. [PMID: 34238121 DOI: 10.1080/15376516.2021.1953204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Acrolein, a known toxin in tobacco smoke, has been demonstrated to be associated with inflammatory cardiovascular diseases, such as atherosclerosis. However, the definite mechanism of acrolein-induced inflammation remains unclear. Here, we report that acrolein induces reactive oxygen species (ROS) production in EAhy926 cells. Additionally, acrolein induces EAhy926 cells' inflammatory response and pyroptosis by activating NOD-like receptor protein 3 (NLRP3) inflammasome. Also, acrolein-induced cytotoxicity could be attenuated by N-acetyl-L-cysteine (NAC). Furthermore, acrolein upregulates the level of autophagy which can be reversed by NAC. Notably, the present study also indicates that autophagy inhibited by inhibitor 3-methyladenine (3MA) and siAtg7 exacerbate acrolein-induced NLRP3 inflammasome activation and pyroptosis. In summary, acrolein induced cytotoxicity by ROS-mediated NLRP3 inflammasome activation, and ROS upregulates the level of autophagy to inhibit the NLRP3 inflammasome excessive activation, indicating the bidirectional role of ROS in acrolein-induced cellular inflammation. Our results may provide novel mechanistic insights into acrolein-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Liping Jiang
- Preventive Medicine Laboratory, College of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Songsong Luo
- Department of Internal Medicine, The Affiliated Zhong Shan Hospital of Dalian University, Dalian, Liaoning, China
| | - Tianming Qiu
- Department of Occupational and Environmental Health, College of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | | | - Chunteng Jiang
- Department of Internal Medicine, The Affiliated Zhong Shan Hospital of Dalian University, Dalian, Liaoning, China
| | - Xiance Sun
- Department of Occupational and Environmental Health, College of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Guang Yang
- Department of Nutrition and Food Safety, College of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Cong Zhang
- Department of Nutrition and Food Safety, College of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaofang Liu
- Department of Nutrition and Food Safety, College of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Lijie Jiang
- Department of Internal Medicine, The Affiliated Zhong Shan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
23
|
Du Y, Guo H, Guo L, Miao J, Ren H, Liu K, Ren L, He J, Wang X, Chen J, Li J, Wang Y, Wang J, Huang N. The regulatory effect of acetylation of HMGN2 and H3K27 on pyocyanin-induced autophagy in macrophages by affecting Ulk1 transcription. J Cell Mol Med 2021; 25:7524-7537. [PMID: 34278675 PMCID: PMC8335688 DOI: 10.1111/jcmm.16788] [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: 01/17/2021] [Revised: 06/05/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022] Open
Abstract
Pyocyanin (PYO) is a major virulence factor secreted by Pseudomonas aeruginosa, and autophagy is a crucial homeostatic mechanism for the interaction between the pathogens and the host. It remains unknown whether PYO leads to autophagy in macrophages by regulating histone acetylation. The high mobility group nucleosomal binding domain 2 (HMGN2) has been reported to regulate the PYO‐induced autophagy and oxidative stress in the epithelial cells; however, the underlying molecular mechanism has not been fully elucidated. In this study, PYO was found to induce autophagy in macrophages, and the mechanism might be correlated with the up‐regulation of HMGN2 acetylation (HMGN2ac) and the down‐regulation of H3K27 acetylation (H3K27ac) by modulation of the activities of acetyltransferases and deacetylases. Moreover, we further demonstrated that the up‐regulated HMGN2ac enhances its recruitment to the Ulk1 promoter, while the down‐regulation of H3K27ac reduces its recruitment to the Ulk1 promoter, thereby promoting or inhibiting the transcription of Ulk1. In conclusion, HMGN2ac and H3K27ac play regulatory roles in the PYO‐induced autophagy in macrophages.
Collapse
Affiliation(s)
- Yu Du
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.,Department of Anesthesiology, Nanchong Central Hospital, Second Clinical Medical Institution, North Sichuan Medical College, Nanchong, China
| | - Hongjun Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijuan Guo
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junming Miao
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hongyu Ren
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Keyun Liu
- Department of Physiology, School of Medicine, Hubei University for Nationalities, Enshi, China
| | - Laibin Ren
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jinchen He
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoying Wang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junli Chen
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jingyu Li
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Wang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Ji Wang
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ning Huang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Fang Y, Peng K. Regulation of innate immune responses by cell death-associated caspases during virus infection. FEBS J 2021; 289:4098-4111. [PMID: 34089572 DOI: 10.1111/febs.16051] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/04/2021] [Accepted: 06/03/2021] [Indexed: 01/04/2023]
Abstract
Viruses are obligate intracellular pathogens that rely on cellular machinery for successful replication and dissemination. The host cells encode a number of different strategies to sense and restrict the invading viral pathogens. Caspase-mediated programmed cell death pathways that are triggered by virus infection, such as apoptosis and pyroptosis, provide a means for the infected cells to limit viral proliferation, leading to suicidal cell death (apoptosis) or lytic cell death and alerting uninfected cells to mount anti-viral responses (pyroptosis). However, some viruses can employ activated caspases to dampen the anti-viral responses and facilitate viral replication through cleavage of critical molecules of the innate immune pathways. The regulation of innate immune responses by caspase activation during virus infection has recently become an important topic. In this review, we briefly introduce the characteristics of different classes of caspases and the cell death pathways regulated by these caspases. We then describe how viruses trigger or dampen caspase activation during infection and how these activated caspases regulate three major innate immune response pathways of viral infections: the retinoic acid-inducible gene I-like receptor, toll-like receptor and cyclic GMP-AMP synthase-stimulator of interferon genes pathways.
Collapse
Affiliation(s)
- Yujie Fang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ke Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
25
|
Al-Omar MS, Jabir M, Karsh E, Kadhim R, Sulaiman GM, Taqi ZJ, Khashan KS, Mohammed HA, Khan RA, Mohammed SAA. Gold Nanoparticles and Graphene Oxide Flakes Enhance Cancer Cells' Phagocytosis through Granzyme-Perforin-Dependent Biomechanism. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1382. [PMID: 34073808 PMCID: PMC8225074 DOI: 10.3390/nano11061382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022]
Abstract
The study aimed to investigate the roles of gold nanoparticles (GNPs) and graphene oxide flakes (GOFs) as phagocytosis enhancers against cancer cells. The nanomaterials were characterized through SEM and UV-VIS absorptions. The GNPs and GOFs increased the macrophages' phagocytosis ability in engulfing, thereby annihilating the cancer cells in both in vitro and in vivo conditions. The GNPs and GOFs augmented serine protease class apoptotic protein, granzyme, passing through the aquaporin class protein, perforin, with mediated delivery through the cell membrane site for the programmed, calibrated, and conditioned cancer cells killing. Additionally, protease inhibitor 3,4-dichloroisocoumarin (DCI) significantly reduced granzyme and perforin activities of macrophages. The results demonstrated that the GOFs and GNPs increased the activation of phagocytic cells as a promising strategy for controlling cancer cells by augmenting the cell mortality through the granzyme-perforin-dependent mechanism.
Collapse
Affiliation(s)
- Mohsen S. Al-Omar
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia; (M.S.A.-O.); (H.A.M.)
- Medicinal Chemistry and Pharmacognosy Department, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Majid Jabir
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq; (E.K.); (R.K.); (Z.J.T.); (K.S.K.)
| | - Esraa Karsh
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq; (E.K.); (R.K.); (Z.J.T.); (K.S.K.)
| | - Rua Kadhim
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq; (E.K.); (R.K.); (Z.J.T.); (K.S.K.)
| | - Ghassan M. Sulaiman
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq; (E.K.); (R.K.); (Z.J.T.); (K.S.K.)
| | - Zainab J. Taqi
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq; (E.K.); (R.K.); (Z.J.T.); (K.S.K.)
| | - Khawla S. Khashan
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq; (E.K.); (R.K.); (Z.J.T.); (K.S.K.)
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia; (M.S.A.-O.); (H.A.M.)
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo 11371, Egypt
| | - Riaz A. Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia; (M.S.A.-O.); (H.A.M.)
| | - Salman A. A. Mohammed
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| |
Collapse
|
26
|
Liao Y, Liu C, Wang J, Song Y, Sabir N, Hussain T, Yao J, Luo L, Wang H, Cui Y, Yang L, Zhao D, Zhou X. Caspase-1 inhibits IFN-β production via cleavage of cGAS during M. bovis infection. Vet Microbiol 2021; 258:109126. [PMID: 34020176 DOI: 10.1016/j.vetmic.2021.109126] [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: 04/25/2020] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
Mycobacterium bovis (M. bovis) infection triggers cytokine production via pattern recognition receptors. These cytokines include type I interferons (IFNs) and interleukin-1β (IL-1β). Excessive type I IFN levels impair host resistance to M. bovis infection. Therefore, strict control of type I IFN production is helpful to reduce pathological damage and bacterial burden. Here, we found that a deficiency in caspase-1, which is the critical component of the inflammasome responsible for IL-1β production, resulted in increased IFN-β production upon M. bovis infection. Subsequent experiments demonstrated that caspase-1 activation reduced cyclic GMP-AMP synthase (cGAS) expression, thereby inhibiting downstream TANK-binding kinase 1 (TBK1)- interferon regulatory factor 3 (IRF3) signaling and ultimately reducing IFN production. A deficiency in caspase-1 activation enhanced the bacterial burden during M. bovis infection in vitro and in vivo and aggravated pathological lesion formation. Thus, caspase-1 activation reduced IFN-β production upon M. bovis infection by dampening cGAS-TBK1-IRF3 signaling, suggesting that the inflammasome protects hosts by negatively regulating harmful cytokines.
Collapse
Affiliation(s)
- Yi Liao
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China; College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, China
| | - Chunfa Liu
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Jie Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Yinjuan Song
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Naveed Sabir
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Tariq Hussain
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Jiao Yao
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Lijia Luo
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Haoran Wang
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Yongyong Cui
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Evanston, IL, 60208, USA
| | - Lifeng Yang
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Deming Zhao
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China
| | - Xiangmei Zhou
- Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100093, China.
| |
Collapse
|
27
|
Autophagy in the HTR-8/SVneo Cell Oxidative Stress Model Is Associated with the NLRP1 Inflammasome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2353504. [PMID: 33854691 PMCID: PMC8019638 DOI: 10.1155/2021/2353504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 11/17/2022]
Abstract
We investigated whether there was activation of NLRP1 inflammasomes and excessive autophagy in oxidative stress damage. And we further demonstrate whether there is a cascade relationship between the activation of NLRP1 inflammasomes and the phenomenon of excessive autophagy. To observe the expression level of the NLRP1 inflammasome group in the pathological process of trophoblast cell oxidative stress, western blot, immunofluorescence, and qRT-PCR were performed. Autophagy in trophoblast cells after the action of H2O2 was detected by using normal trophoblast cells' NLRP1-specific activator (MDP) as a positive control. The presence of excessive autophagy was determined by comparing it with the autophagy-related proteins in normal trophoblast cells. Through siRNA-NLRP1, we investigated the role of oxidative stress and the NLRP1 inflammasome in autophagy in cells. 100 μmol MDP for 24 hours can be used as the optimal concentration of the NLRP1 activator. In human placental trophoblast oxidative stress, the model group significantly increased the expression level of inflammasome IL-1β, CASP1, and NLRP1, compared with the control group NLRP3, and LC3-II, Beclin-1, ATG5, ATG7, and p62 overactivated the autophagy ability of cells. After the activation of NLRP1, the expression of these inflammasomes increased, accompanied by the decrease in autophagy. After the expression of NLRP1 was silenced by RNAi, the expression of inflammasome IL-1β, CASP1, and NLRP3 was also decreased. Still, the autophagy level was increased, which was manifested by the high expression of LC3-II, Beclin-1, ATG5, and ATG7 and the decrease in p62. Trophoblast cells showed the expression of NLRP1 protein and excessive autophagy under oxidative stress. Simultaneously, the NLRP1 inflammasome of trophoblast cells in the state of oxidative stress was correlated with autophagy. Inflammasome activation and autophagy were shown to be linked and to influence each other mutually. These may also provide new therapeutic targets in a pathological pregnancy.
Collapse
|
28
|
The NLRP3 Inflammasome and Its Role in the Pathogenicity of Leukemia. Int J Mol Sci 2021; 22:ijms22031271. [PMID: 33525345 PMCID: PMC7865748 DOI: 10.3390/ijms22031271] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation contributes to the development and progression of various tumors. Especially where the inflammation is mediated by cells of the innate immune system, the NLRP3 inflammasome plays an important role, as it senses and responds to a variety of exogenous and endogenous pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). The NLRP3 inflammasome is responsible for the maturation and secretion of the proinflammatory cytokines interleukin-1β (IL-1β) and IL-18 and for the induction of a type of inflammatory cell death known as pyroptosis. Overactivation of the NLRP3 inflammasome can be a driver of various diseases. Since leukemia is known to be an inflammation-driven cancer and IL-1β is produced in elevated levels by leukemic cells, research on NLRP3 in the context of leukemia has increased in recent years. In this review, we summarize the current knowledge on leukemia-promoting inflammation and, in particular, the role of the NLRP3 inflammasome in different types of leukemia. Furthermore, we examine a connection between NLRP3, autophagy and leukemia.
Collapse
|
29
|
Biasizzo M, Kopitar-Jerala N. Interplay Between NLRP3 Inflammasome and Autophagy. Front Immunol 2020; 11:591803. [PMID: 33163006 PMCID: PMC7583715 DOI: 10.3389/fimmu.2020.591803] [Citation(s) in RCA: 324] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
The NLRP3 inflammasome is cytosolic multi-protein complex that induces inflammation and pyroptotic cell death in response to both pathogen (PAMPs) and endogenous activators (DAMPs). Recognition of PAMPs or DAMPs leads to formation of the inflammasome complex, which results in activation of caspase-1, followed by cleavage and release of pro-inflammatory cytokines. Excessive activation of NLRP3 inflammasome can contribute to development of inflammatory diseases and cancer. Autophagy is vital intracellular process for recycling and removal of damaged proteins and organelles, as well as destruction of intracellular pathogens. Cytosolic components are sequestered in a double-membrane vesicle-autophagosome, which then fuses with lysosome resulting in degradation of the cargo. The autophagy dysfunction can lead to diseases with hyperinflammation and excessive activation of NLRP3 inflammasome and thus acts as a major regulator of inflammasomes. Autophagic removal of NLRP3 inflammasome activators, such as intracellular DAMPs, NLRP3 inflammasome components, and cytokines can reduce inflammasome activation and inflammatory response. Likewise, inflammasome signaling pathways can regulate autophagic process necessary for balance between required host defense inflammatory response and prevention of excessive and detrimental inflammation. Autophagy has a protective role in some inflammatory diseases associated with NLRP3 inflammasome, including gouty arthritis, familial Mediterranean fever (FMF), and sepsis. Understanding the interregulation between these two essential biological processes is necessary to comprehend the biological mechanisms and designing possible treatments for multiple inflammatory diseases.
Collapse
Affiliation(s)
- Monika Biasizzo
- Department of Biochemistry, Molecular and Structural Biology, JoŽef Stefan Institute, Ljubljana, Slovenia.,Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Nataša Kopitar-Jerala
- Department of Biochemistry, Molecular and Structural Biology, JoŽef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
30
|
Cavinato L, Genise E, Luly FR, Di Domenico EG, Del Porto P, Ascenzioni F. Escaping the Phagocytic Oxidative Burst: The Role of SODB in the Survival of Pseudomonas aeruginosa Within Macrophages. Front Microbiol 2020; 11:326. [PMID: 32210934 PMCID: PMC7077434 DOI: 10.3389/fmicb.2020.00326] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/14/2020] [Indexed: 12/25/2022] Open
Abstract
Reactive oxygen species (ROS) are small oxygen-derived molecules that are used to control infections by phagocytic cells. In macrophages, the oxidative burst produced by the NOX2 NADPH-oxidase is essential to eradicate engulfed pathogens by both oxidative and non-oxidative killing. Indeed, while the superoxide anion (O2-) produced by NOX2, and the other ROS derived from its transformation, can directly target pathogens, ROS also contribute to activation of non-oxidative microbicidal effectors. The response of pathogens to the phagocytic oxidative burst includes the expression of different enzymes that target ROS to reduce their toxicity. Superoxide dismutases (SODs) are the primary scavengers of O2-, which is transformed into H2O2. In the Gram-negative Salmonella typhimurium, periplasmic SODCI has a major role in bacterial resistance to NOX-mediated oxidative stress. In Pseudomonas aeruginosa, the two periplasmic SODs, SODB, and SODM, appear to contribute to bacterial virulence in small-animal models. Furthermore, NOX2 oxidative stress is essential to restrict P. aeruginosa survival in macrophages early after infection. Here, we focused on the role of P. aeruginosa SODs in the counteracting of the lethal effects of the macrophage oxidative burst. Through this study of the survival of sod mutants in macrophages and the measurement of ROS in infected macrophages, we have identified a dual, antagonistic, role for SODB in P. aeruginosa survival. Indeed, the survival of the sodB mutants, but not of the sodM mutants, was greater than that of the wild-type (WT) bacteria early after infection, and sodB-infected macrophages showed higher levels of O2- and lower levels of H2O2. This suggests that SODB contributes to the production of lethal doses of H2O2 within the phagosome. However, later on following infection, the sodB mutants survived less that the WT bacteria, which highlights the pro-survival role of SODB. We have explained this defensive role through an investigation of the activation of autophagy, which was greater in the sodB-infected macrophages.
Collapse
Affiliation(s)
- Luca Cavinato
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Elena Genise
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Francesco R Luly
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Enea G Di Domenico
- Microbiology and Virology, San Gallicano Dermatologic Institute, IRCCS, Rome, Italy
| | - Paola Del Porto
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Fiorentina Ascenzioni
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
31
|
Cho DH, Kim JK, Jo EK. Mitophagy and Innate Immunity in Infection. Mol Cells 2020; 43:10-22. [PMID: 31999918 PMCID: PMC6999710 DOI: 10.14348/molcells.2020.2329] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 02/08/2023] Open
Abstract
Mitochondria have several quality control mechanisms by which they maintain cellular homeostasis and ensure that the molecular machinery is protected from stress. Mitophagy, selective autophagy of mitochondria, promotes mitochondrial quality control by inducing clearance of damaged mitochondria via the autophagic machinery. Accumulating evidence suggests that mitophagy is modulated by various microbial components in an attempt to affect the innate immune response to infection. In addition, mitophagy plays a key role in the regulation of inflammatory signaling, and mitochondrial danger signals such as mitochondrial DNA translocated into the cytosol can lead to exaggerated inflammatory responses. In this review, we present current knowledge on the functional aspects of mitophagy and its crosstalk with innate immune signaling during infection. A deeper understanding of the role of mitophagy could facilitate the development of more effective therapeutic strategies against various infections.
Collapse
Affiliation(s)
- Dong-Hyung Cho
- School of Life Sciences, Kyungpook National University, Daegu 41566,
Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015,
Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015,
Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015,
Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015,
Korea
| |
Collapse
|
32
|
Chen S, Deng Y, He Q, Chen Y, Wang D, Sun W, He Y, Zou Z, Liang Z, Chen R, Yao L, Tao A. Toll-like Receptor 4 Deficiency Aggravates Airway Hyperresponsiveness and Inflammation by Impairing Neutrophil Apoptosis in a Toluene Diisocyanate-Induced Murine Asthma Model. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:608-625. [PMID: 32400128 PMCID: PMC7225000 DOI: 10.4168/aair.2020.12.4.608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/28/2019] [Accepted: 01/01/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Accumulating evidence has suggested that toll-like receptor 4 (TLR4) is critically involved in the pathogenesis of asthma. The aim of this study was to investigate the role of TLR4 in toluene diisocyanate (TDI)-induced allergic airway inflammation. METHODS TLR4-/- and wild-type (WT) C57BL/10J mice were sensitized and challenged with TDI to generate a TDI-induced asthma model. B-cell lymphoma 2 (Bcl-2) inhibitors, ABT-199 (4 mg/kg) and ABT-737 (4 mg/kg), were intranasally given to TDI-exposed TLR4-/- mice after each challenge. RESULTS TDI exposure led to increased airway hyperresponsiveness (AHR), granulocyte flux, bronchial epithelial shedding and extensive submucosal collagen deposition, which were unexpectedly aggravated by TLR4 deficiency. Following TDI challenge, TLR4-/- mice exhibited down-regulated interleukin-17A and increased colony-stimulating factor 3 in bronchoalveolar lavage fluid (BALF), while WT mice did not. In addition, TLR4 deficiency robustly suppressed the expression of NOD-like receptor family pyrin domain containing 3 and NLR family CARD domain containing 4, decreased caspase-1 activity in TDI-exposed mice, but had no effect on the level of high mobility group box 1 in BALF. Flow cytometry revealed that TDI hampered both neutrophil and eosinophil apoptosis, of which neutrophil apoptosis was further inhibited in TDI-exposed TLR4-/- mice, with marked up-regulation of Bcl-2. Moreover, inhibition of Bcl-2 with either ABT-199 or ABT-737 significantly alleviated neutrophil recruitment by promoting apoptosis. CONCLUSIONS These data indicated that TLR4 deficiency promoted neutrophil infiltration by impairing its apoptosis via up-regulation of Bcl-2, thereby resulting in deteriorated AHR and airway inflammation, which suggests that TLR4 could be a negative regulator of TDI-induced neutrophilic inflammation.
Collapse
Affiliation(s)
- Shuyu Chen
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China
| | - Yao Deng
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China
| | - Qiaoling He
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China
| | - Yanbo Chen
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - De Wang
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China
| | - Weimin Sun
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China
| | - Ying He
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China
| | - Zehong Zou
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhenyu Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Rongchang Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lihong Yao
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Ailin Tao
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.,Center for Immunology, Inflammation & Immune-Mediated Disease, Guangzhou Medical University, Guangzhou, China. ,
| |
Collapse
|
33
|
Xia B, Yu J, He T, Liu X, Su J, Wang M, Wang J, Zhu Y. Lactobacillus johnsonii L531 ameliorates enteritis via elimination of damaged mitochondria and suppression of SQSTM1-dependent mitophagy in a Salmonella infantis model of piglet diarrhea. FASEB J 2019; 34:2821-2839. [PMID: 31908018 DOI: 10.1096/fj.201901445rrr] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 01/04/2023]
Abstract
Newly weaned piglets challenged with Salmonella infantis were particularly susceptible, whereas oral preadministration of Lactobacillus johnsonii L531 alleviated enteritis and promoted intestinal secretory IgA production. Salmonella infantis-induced activation of NLRC4 and NLRP3 inflammasomes and (nuclear factor kappa B) NF-κB signaling in the small intestine was also inhibited by L. johnsonii L531 pretreatment, thus limiting inflammation. An IPEC-J2 cell model of S. infantis infection yielded similar results. Salmonella infantis infection also resulted in mitochondrial damage and impaired mitophagy in the ileum and IPEC-J2 cells, as demonstrated by immunofluorescence colocalization of mitochondria with microtubule-binding protein light chain 3 (LC3) and high expression of autophagy-related proteins PTEN-induced putative kinase 1 (PINK1), sequestosome 1 (SQSTM1/p62), optineurin (OPTN), and LC3 by Western blotting analysis. However, L. johnsonii L531 pretreatment reduced both the extent of mitochondrial damage and autophagy-related protein expression. Our findings suggest that the amelioration of S. infantis-associated enteritis by L. johnsonii L531 is associated with regulation of NLRC4 and NLRP3 inflammasomes and NF-κB signaling pathway activation and suppression of mitochondrial damage. Amelioration of impaired mitophagy by L. johnsonii L531 could involve eliminating damaged mitochondria and regulating S. infantis-induced activation of the NF-κB-SQSTM1mitophagy signaling pathway in host cells to prevent the further mitochondrial damage and S. infantis dissemination.
Collapse
Affiliation(s)
- Bing Xia
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiao Yu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ting He
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiao Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jinhui Su
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Meijun Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiufeng Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yaohong Zhu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
34
|
Apoptotic Caspases Suppress Type I Interferon Production via the Cleavage of cGAS, MAVS, and IRF3. Mol Cell 2019; 74:19-31.e7. [PMID: 30878284 DOI: 10.1016/j.molcel.2019.02.013] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/13/2018] [Accepted: 02/08/2019] [Indexed: 01/12/2023]
Abstract
Viral infection triggers host defenses through pattern-recognition receptor-mediated cytokine production, inflammasome activation, and apoptosis of the infected cells. Inflammasome-activated caspases are known to cleave cyclic GMP-AMP synthase (cGAS). Here, we found that apoptotic caspases are critically involved in regulating both DNA and RNA virus-triggered host defenses, in which activated caspase-3 cleaved cGAS, MAVS, and IRF3 to prevent cytokine overproduction. Caspase-3 was exclusively required in human cells, whereas caspase-7 was involved only in murine cells to inactivate cGAS, reflecting distinct regulatory mechanisms in different species. Caspase-mediated cGAS cleavage was enhanced in the presence of dsDNA. Alternative MAVS cleavage sites were used to ensure the inactivation of this critical protein. Elevated type I IFNs were detected in caspase-3-deficient cells without any infection. Casp3-/- mice consistently showed increased resistance to viral infection and experimental autoimmune encephalomyelitis. Our results demonstrate that apoptotic caspases control innate immunity and maintain immune homeostasis against viral infection.
Collapse
|
35
|
Miyamoto S. Autophagy and cardiac aging. Cell Death Differ 2019; 26:653-664. [PMID: 30692640 PMCID: PMC6460392 DOI: 10.1038/s41418-019-0286-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/02/2019] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death and the prevalence of CVD dramatically increases with age. Cardiac aging is associated with hypertrophy, fibrosis, inflammation, and decreased contractility. Autophagy, a bulk degradation/recycling system, is essential to maintain cellular homeostasis. Cardiac autophagy is decreased with age, and misfolded proteins and dysfunctional mitochondria are accumulated in the aging heart. Inhibition of autophagy leads to exacerbated cardiac aging, while stimulation of autophagy improves cardiac function and also increases lifespan in many organisms. Thus autophagy represents a potential therapeutic target for aging-related cardiac dysfunction. This review discusses recent progress in our understanding of the role and regulation of autophagy in the aging heart.
Collapse
Affiliation(s)
- Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
36
|
Yingchun L, Huihan W, Rong Z, Guojun Z, Ying Y, Zhuogang L. Antitumor Activity of Asiaticoside Against Multiple Myeloma Drug-Resistant Cancer Cells Is Mediated by Autophagy Induction, Activation of Effector Caspases, and Inhibition of Cell Migration, Invasion, and STAT-3 Signaling Pathway. Med Sci Monit 2019; 25:1355-1361. [PMID: 30785126 PMCID: PMC6391856 DOI: 10.12659/msm.913397] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Accumulating evidence suggests that plant-derived molecules may prove extremely beneficial in the development of chemotherapy for deadly cancer types. Multiple myeloma is a rare and incurable type of cancers. Very little research has been directed towards the development of chemotherapy for the management of multiple myeloma. Here, the anticancer effects of a plant-derived triterpenoid, Asiaticoside, were examined against the drug-resistant myeloma cell line KM3/BTZ. Material/Methods Cell viability was determined by CCK-8 assay and autophagy was checked by transmission electron microscopy. ROS levels were determined by flow cytometery. Cell migration and invasion were examined by Transwell assay. Protein expression was assessed by Western blotting. Results The results showed that Asiaticoside inhibits the growth of the KM3/BTZ cells and exhibited an IC50 of 12 μM. Further, it was observed that the anticancer effects of Asiaticoside are due to the induction of autophagy allied with upsurge of the expression of LC3-II. Moreover, the expression of the effector caspases in the KM3/BTZ cells was also altered. Asiaticoside also caused accretion of the ROS in the KM3/BTZ cells and inhibited their migratory and invasive properties via modulation of the STAT-3 signaling pathway. Conclusions Asiaticoside may prove useful in the management and treatment of the multiple myeloma and needs further investigation.
Collapse
Affiliation(s)
- Li Yingchun
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Wang Huihan
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Zhang Rong
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Zhang Guojun
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yang Ying
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Liu Zhuogang
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
37
|
Protective Features of Autophagy in Pulmonary Infection and Inflammatory Diseases. Cells 2019; 8:cells8020123. [PMID: 30717487 PMCID: PMC6406971 DOI: 10.3390/cells8020123] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process involving autolysosomal degradation of cellular components, including protein aggregates, damaged organelles (such as mitochondria, endoplasmic reticulum, and others), as well as various pathogens. Thus, the autophagy pathway represents a major adaptive response for the maintenance of cellular and tissue homeostasis in response to numerous cellular stressors. A growing body of evidence suggests that autophagy is closely associated with diverse human diseases. Specifically, acute lung injury (ALI) and inflammatory responses caused by bacterial infection or xenobiotic inhalation (e.g., chlorine and cigarette smoke) have been reported to involve a spectrum of alterations in autophagy phenotypes. The role of autophagy in pulmonary infection and inflammatory diseases could be protective or harmful dependent on the conditions. In this review, we describe recent advances regarding the protective features of autophagy in pulmonary diseases, with a focus on ALI, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), tuberculosis, pulmonary arterial hypertension (PAH) and cystic fibrosis.
Collapse
|
38
|
Yabal M, Calleja DJ, Simpson DS, Lawlor KE. Stressing out the mitochondria: Mechanistic insights into NLRP3 inflammasome activation. J Leukoc Biol 2018; 105:377-399. [PMID: 30589456 DOI: 10.1002/jlb.mr0318-124r] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022] Open
Abstract
Inflammasomes are multimeric protein complexes that induce the cleavage and release of bioactive IL-1β and cause a lytic form of cell death, termed pyroptosis. Due to its diverse triggers, ranging from infectious pathogens and host danger molecules to environmental irritants, the NOD-like receptor protein 3 (NLRP3) inflammasome remains the most widely studied inflammasome to date. Despite intense scrutiny, a universal mechanism for its activation remains elusive, although, recent research has focused on mitochondrial dysfunction or potassium (K+ ) efflux as key events. In this review, we give a general overview of NLRP3 inflammasome activation and explore the recently emerging noncanonical and alternative pathways to NLRP3 activation. We highlight the role of the NLRP3 inflammasome in the pathogenesis of metabolic disease that is associated with mitochondrial and oxidative stress. Finally, we interrogate the mechanisms proposed to trigger NLRP3 inflammasome assembly and activation. A greater understanding of how NLRP3 inflammasome activation is triggered may reveal new therapeutic targets for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Monica Yabal
- III. Medical Department for Hematology and Oncology, Kinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Dale J Calleja
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Daniel S Simpson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Kate E Lawlor
- Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
39
|
Rawat P, Teodorof-Diedrich C, Spector SA. Human immunodeficiency virus Type-1 single-stranded RNA activates the NLRP3 inflammasome and impairs autophagic clearance of damaged mitochondria in human microglia. Glia 2018; 67:802-824. [PMID: 30582668 DOI: 10.1002/glia.23568] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022]
Abstract
Despite the availability of antiretroviral therapy (ART) that fully suppresses human immunodeficiency virus type-1 (HIV), markers of inflammation and minor neurocognitive impairment are frequently identified in HIV-infected persons. Increasing data support that low-level replication defective viral RNA is made by infected cells despite the absence of infectious virus. Specific GU-rich single-stranded RNA from the HIV long terminal repeat region (ssRNA40) signaling through toll-like receptor (TLR)-7 and -8 has been shown to induce the secretion of interleukin-1β (IL-1β) in primary monocytes. Here, we examined the activation of microglial cells by HIV ssRNA40 and the potential subsequent neurotoxicity. Our findings show that exposure of human primary microglia to ssRNA40 activates the NLR family pyrin domain containing 3 (NLRP3) inflammasome. Following exposure to ssRNA40, pro-inflammatory cytokines IL-1β, IL-18, and neurotoxic cytokines TNF-α, IL-1α, and C1q expression and extracellular secretion are increased. The released cytokines are functional since culture supernatants from ssRNA40 exposed microglia-induced toxicity of human primary neurons. Moreover, inflammasome activation of microglia increased ROS generation with a loss of mitochondrial membrane potential and mitochondrial integrity. Treatment with ssRNA40 resulted in a blockade of autophagy/mitophagy mediated negative regulation of NLRP3 inflammasome activity with the release of inflammatory cytokines, caspase-1 activation, and pyroptotic microglial cell death. Thus, HIV ssRNA mediated activation of microglial cells can contribute to neurotoxicity and neurodegeneration via secretion of inflammatory and neurotoxic cytokines. These findings provide a potential mechanism that explains the frequent minor cognitive deficits and chronic inflammation that persist in HIV-infected persons despite treatment with suppressive ART.
Collapse
Affiliation(s)
- Pratima Rawat
- Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, California
| | - Carmen Teodorof-Diedrich
- Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, California
| | - Stephen A Spector
- Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, California.,Rady Children's Hospital, San Diego, California
| |
Collapse
|
40
|
Perkins DJ, Richard K, Hansen AM, Lai W, Nallar S, Koller B, Vogel SN. Autocrine-paracrine prostaglandin E 2 signaling restricts TLR4 internalization and TRIF signaling. Nat Immunol 2018; 19:1309-1318. [PMID: 30397349 PMCID: PMC6240378 DOI: 10.1038/s41590-018-0243-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/14/2018] [Indexed: 12/12/2022]
Abstract
The unique cell biology of Toll-like receptor 4 (TLR4) allows it to initiate two signal transduction cascades: a Mal (TIRAP)–MyD88-dependent signal from the cell surface that regulates proinflammatory cytokines and a TRAM–TRIF-dependent signal from endosomes that drives type I interferon production. Negative feedback circuits to limit TLR4 signals from both locations are necessary to balance the inflammatory response. We describe a negative feedback loop driven by autocrine-paracrine prostaglandin E2 (PGE2), and the PGE2 receptor, EP4, which restricted TRIF-dependent signals and IFN-β induction through regulation of TLR4 trafficking. Inhibition of PGE2 production or EP4 antagonism increased the rate of TLR4 endosomal translocation, and amplified TRIF-dependent IRF3 and caspase 8 activation. This PGE2-driven mechanism restricted TLR4-TRIF signaling in vitro upon infection of macrophages by Gram-negative pathogens Escherichia coli and Citrobacter rodentium and protected mice against Salmonella enteritidis serovar Typhimurium (ST)-induced mortality. Thus, PGE2 restricts TLR4-TRIF signaling specifically in response to lipopolysaccharide.
Collapse
Affiliation(s)
- Darren J Perkins
- Department of Microbiology and Immunology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD, USA.
| | - Katharina Richard
- Department of Microbiology and Immunology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD, USA
| | - Anne-Marie Hansen
- Department of Microbiology and Immunology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD, USA
| | - Wendy Lai
- Department of Microbiology and Immunology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD, USA
| | - Shreeram Nallar
- Department of Microbiology and Immunology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD, USA
| | - Beverly Koller
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland, Baltimore, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
41
|
Zheng Y, Liu Q, Wu Y, Ma L, Zhang Z, Liu T, Jin S, She Y, Li YP, Cui J. Zika virus elicits inflammation to evade antiviral response by cleaving cGAS via NS1-caspase-1 axis. EMBO J 2018; 37:embj.201899347. [PMID: 30065070 DOI: 10.15252/embj.201899347] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 01/02/2023] Open
Abstract
Viral infection triggers host innate immune responses, which primarily include the activation of type I interferon (IFN) signaling and inflammasomes. Here, we report that Zika virus (ZIKV) infection triggers NLRP3 inflammasome activation, which is further enhanced by viral non-structural protein NS1 to benefit its replication. NS1 recruits the host deubiquitinase USP8 to cleave K11-linked poly-ubiquitin chains from caspase-1 at Lys134, thus inhibiting the proteasomal degradation of caspase-1. The enhanced stabilization of caspase-1 by NS1 promotes the cleavage of cGAS, which recognizes mitochondrial DNA release and initiates type I IFN signaling during ZIKV infection. NLRP3 deficiency increases type I IFN production and strengthens host resistance to ZIKVin vitro and in vivo Taken together, our work unravels a novel antagonistic mechanism employed by ZIKV to suppress host immune response by manipulating the interplay between inflammasome and type I IFN signaling, which might guide the rational design of therapeutics in the future.
Collapse
Affiliation(s)
- Yanyan Zheng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qingxiang Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yaoxing Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ling Ma
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Zhang
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tao Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuanchu She
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yi-Ping Li
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China .,Department of Infectious Disease, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
42
|
The mitochondrial protease HtrA2 restricts the NLRP3 and AIM2 inflammasomes. Sci Rep 2018; 8:8446. [PMID: 29855523 PMCID: PMC5981608 DOI: 10.1038/s41598-018-26603-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
Activation of the inflammasome pathway is crucial for effective intracellular host defense. The mitochondrial network plays an important role in inflammasome regulation but the mechanisms linking mitochondrial homeostasis to attenuation of inflammasome activation are not fully understood. Here, we report that the Parkinson’s disease-associated mitochondrial serine protease HtrA2 restricts the activation of ASC-dependent NLRP3 and AIM2 inflammasomes, in a protease activity-dependent manner. Consistently, disruption of the protease activity of HtrA2 results in exacerbated NLRP3 and AIM2 inflammasome responses in macrophages ex vivo and systemically in vivo. Mechanistically, we show that the HtrA2 protease activity regulates autophagy and controls the magnitude and duration of inflammasome signaling by preventing prolonged accumulation of the inflammasome adaptor ASC. Our findings identify HtrA2 as a non-redundant mitochondrial quality control effector that keeps NLRP3 and AIM2 inflammasomes in check.
Collapse
|
43
|
Lai M, Yao H, Shah SZA, Wu W, Wang D, Zhao Y, Wang L, Zhou X, Zhao D, Yang L. The NLRP3-Caspase 1 Inflammasome Negatively Regulates Autophagy via TLR4-TRIF in Prion Peptide-Infected Microglia. Front Aging Neurosci 2018; 10:116. [PMID: 29720937 PMCID: PMC5915529 DOI: 10.3389/fnagi.2018.00116] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/03/2018] [Indexed: 12/15/2022] Open
Abstract
Prion diseases are neurodegenerative disorders characterized by the accumulation of misfolded prion protein, spongiform changes in the brain, and brain inflammation as a result of the wide-spread activation of microglia. Autophagy is a highly conserved catabolic process for the clearance of cytoplasmic components, including protein aggregates and damaged organelles; this process also eliminates pathological PrPSc as it accumulates during prion infection. The NALP3 inflammasome is a multiprotein complex that is a component of the innate immune system and is responsible for the release of pro-inflammatory cytokines. Our previous study showed that the neurotoxic prion peptide PrP106-126 induces NALP3 inflammasome activation and subsequent IL-1β release in microglia. Autophagy is involved in the regulation of the immune responses and inflammation in many diseases including neurodegenerative diseases. However, the relationship between autophagy and NALP3 inflammasome in prion diseases has not been investigated. In this study, we demonstrated that the processing and release of mature IL-1β is significantly enhanced by the inhibition of autophagy. Conversely, gene-silencing of the NALP3 inflammasome promotes autophagy. Suppression of TRIF or TLR4 by siRNA attenuated PrP106-126-induced autophagy, which is indicating that the TLR4-TRIF signaling pathway is involved in PrP106-26-induced autophagy. Caspase 1 directly cleaved TRIF to diminish TLR-4-TRIF mediated autophagy. Our findings suggest that the inhibition of autophagy by NALP3 inflammasome is probably mediated by activated Caspase-1-induced TRIF cleavage. This is the first study reporting that the NALP3 inflammasome complex negatively regulates autophagy in response to PrP106-126 stimulation in microglia, and partly explains the mechanism of autophagy inhibition by Caspase-1 in PrP106-126-induced BV2 cell activation. Our findings suggest that autophagy up-regulation and inhibition of Caspase-1 may protect against prion-induced neuroinflammation and accelerate misfolded protein degradation and are potential therapeutic approaches for prion diseases.
Collapse
Affiliation(s)
- Mengyu Lai
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Hao Yao
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Syed Zahid Ali Shah
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Wei Wu
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Di Wang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Ying Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Lu Wang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Deming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Lifeng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, China Agricultural University, Beijing, China
| |
Collapse
|
44
|
lincRNA-Cox2 regulates NLRP3 inflammasome and autophagy mediated neuroinflammation. Cell Death Differ 2018; 26:130-145. [PMID: 29666475 PMCID: PMC6294802 DOI: 10.1038/s41418-018-0105-8] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/02/2018] [Accepted: 03/12/2018] [Indexed: 01/08/2023] Open
Abstract
Inflammasome activation plays key roles in host defense, but also contributes to the pathogenesis of auto-inflammatory, and neurodegenerative diseases. As autophagy is connected with both the innate and adaptive immune systems, autophagic dysfunction is also closely related to inflammation, infection, and neurodegeneration. Here we identify that lincRNA-Cox2, previously known as a mediator of both the activation and repression of immune genes expression in innate immune cells, could bind NF-κB p65 and promote its nuclear translocation and transcription, modulating the expression of inflammasome sensor NLRP3 and adaptor ASC. Knockdown of lincRNA-Cox2 inhibited the inflammasome activation and prevented the lincRNA-Cox2-triggered caspase-1 activation, leading to decreased IL-1β secretion and weakened TIR-domain-containing adapter-inducing interferon-β (TRIF) cleavage, thereby enhancing TRIF-mediated autophagy. Elucidation of the link between lincRNA-Cox2 and the inflammasome-autophagy crosstalk in macrophage and microglia reveals a role for lncRNAs in activation of NLRP3 inflammasome and autophagy, and provides new opportunities for therapeutic intervention in neuroinflammation-dependent diseases.
Collapse
|
45
|
Elevated prostaglandin E 2 post-bone marrow transplant mediates interleukin-1β-related lung injury. Mucosal Immunol 2018; 11:319-332. [PMID: 28589946 PMCID: PMC5720939 DOI: 10.1038/mi.2017.51] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/25/2017] [Indexed: 02/04/2023]
Abstract
Hematopoietic stem cell transplant (HSCT) treats or cures a variety of hematological and inherited disorders. Unfortunately, patients who undergo HSCT are susceptible to infections by a wide array of opportunistic pathogens. Pseudomonas aeruginosa bacteria can have life-threatening effects in HSCT patients by causing lung pathology that has been linked to high levels of the potent pro-inflammatory cytokine, interleukin-1β (IL-1β). Using a murine bone marrow transplant (BMT) model, we show that overexpression of prostaglandin E2 (PGE2) post-BMT signals via EP2 or EP4 to induce cyclic adenosine monophosphate (cAMP), which activates protein kinase A or the exchange protein activated by cAMP (Epac) to induce cAMP response element binding-dependent transcription of IL-1β leading to exacerbated lung injury in BMT mice. Induction of IL-1β by PGE2 is time and dose dependent. Interestingly, IL-1β processing post-P. aeruginosa infection occurs via the enzymatic activity of either caspase-1 or caspase-8. Furthermore, PGE2 can limit autophagy-mediated killing of P. aeruginosa in alveolar macrophages, yet autophagy does not have a role in PGE2-mediated upregulation of IL-1β. Reducing PGE2 levels with indomethacin improved bacterial clearance and reduced IL-1β-mediated acute lung injury in P. aeruginosa-infected BMT mice.
Collapse
|
46
|
Pecenková T, Markovic V, Sabol P, Kulich I, Žárský V. Exocyst and autophagy-related membrane trafficking in plants. JOURNAL OF EXPERIMENTAL BOTANY 2017; 69:47-57. [PMID: 29069430 DOI: 10.1093/jxb/erx363] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Endomembrane traffic in eukaryotic cells functions partially as a means of communication; delivery of membrane in one direction has to be balanced with a reduction at the other end. This effect is typically the case during the defence against pathogens. To combat pathogens, cellular growth and differentiation are suppressed, while endomembrane traffic is poised towards limiting the pathogen attack. The octameric exocyst vesicle-tethering complex was originally discovered as a factor facilitating vesicle-targeting and vesicle-plasma membrane (PM) fusion during exocytosis prior to and possibly during SNARE complex formation. Interestingly, it was recently implicated both in animals and plants in autophagy membrane traffic. In animal cells, the exocyst is integrated into the mTOR-regulated energy metabolism stress/starvation pathway, participating in the formation and especially initiation of an autophagosome. In plants, the first functional link was to autophagy-related anthocyanin import to the vacuole and to starvation. In this concise review, we summarize the current knowledge of exocyst functions in autophagy and defence in plants that might involve unconventional secretion and compare it with animal conditions. Formation of different exocyst complexes during undisturbed cell growth, as opposed to periods of cellular stress reactions involving autophagy, might contribute to the coordination of endomembrane trafficking pathways.
Collapse
Affiliation(s)
- Tamara Pecenková
- Laboratory of Cell Biology, Institute of Experimental Botany, Academy of Sciences of the Czech Republic, Czech Republic
- Laboratory of Cell Morphogenesis, Department of Experimental Plant Biology, Charles University in Prague, Faculty of Science, Czech Republic
| | - Vedrana Markovic
- Laboratory of Cell Biology, Institute of Experimental Botany, Academy of Sciences of the Czech Republic, Czech Republic
- Laboratory of Cell Morphogenesis, Department of Experimental Plant Biology, Charles University in Prague, Faculty of Science, Czech Republic
| | - Peter Sabol
- Laboratory of Cell Morphogenesis, Department of Experimental Plant Biology, Charles University in Prague, Faculty of Science, Czech Republic
| | - Ivan Kulich
- Laboratory of Cell Morphogenesis, Department of Experimental Plant Biology, Charles University in Prague, Faculty of Science, Czech Republic
| | - Viktor Žárský
- Laboratory of Cell Biology, Institute of Experimental Botany, Academy of Sciences of the Czech Republic, Czech Republic
- Laboratory of Cell Morphogenesis, Department of Experimental Plant Biology, Charles University in Prague, Faculty of Science, Czech Republic
| |
Collapse
|
47
|
Ortmann W, Kolaczkowska E. Age is the work of art? Impact of neutrophil and organism age on neutrophil extracellular trap formation. Cell Tissue Res 2017; 371:473-488. [PMID: 29250748 PMCID: PMC5820386 DOI: 10.1007/s00441-017-2751-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022]
Abstract
Neutrophil extracellular traps or NETs are released by highly activated neutrophils in response to infectious agents, sterile inflammation, autoimmune stimuli and cancer. In the cells, the nuclear envelop disintegrates and decondensation of chromatin occurs that depends on peptidylarginine deiminase 4 (PAD4) and neutrophil elastase (NE). Subsequently, proteins from neutrophil granules (e.g., NE, lactoferrin and myeloperoxidase) and the nucleus (histones) bind to decondensed DNA and the whole structure is ejected from the cell. The DNA decorated with potent antimicrobials and proteases can act to contain dissemination of infection and in sterile inflammation NETs were shown to degrade cytokines and chemokines via serine proteases. On the other hand, overproduction of NETs, or their inadequate removal and prolonged presence in vasculature or tissues, can lead to bystander damage or even initiation of diseases. Considering the pros and cons of NET formation, it is of relevance if the stage of neutrophil maturation (immature, mature and senescent cells) affects the capacity to produce NETs as the cells of different age-related phenotypes dominate in given (pathological) conditions. Moreover, the immune system of neonates and elderly individuals is weaker than in adulthood. Is the same pattern followed when it comes to NETs? The overall importance of individual and neutrophil age on the capacity to release NETs is reviewed in detail and the significance of these facts is discussed.
Collapse
Affiliation(s)
- Weronika Ortmann
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Jagiellonian University, ul. Gronostajowa 9, 30-387, Krakow, Poland
| | - Elzbieta Kolaczkowska
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Jagiellonian University, ul. Gronostajowa 9, 30-387, Krakow, Poland.
| |
Collapse
|
48
|
Iraqi propolis increases degradation of IL-1β and NLRC4 by autophagy following Pseudomonas aeruginosa infection. Microbes Infect 2017; 20:89-100. [PMID: 29104144 DOI: 10.1016/j.micinf.2017.10.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/13/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022]
Abstract
Autophagy is a cellular process for maintaining cellular homeostasis. This process can be induced by different factors, such as immune stimuli and pathogen-associated molecules. Autophagy has an important role in the control of IL-1β secretion by macrophages and other cell types. In present study, we describe a novel role for Iraqi propolis affecting autophagy in controlling the secretion of IL-1β in bone-marrow macrophages (BMDMs). After infection with Pseudomonas aeruginosa in the presence of propolis, the degradation of IL-1β was induced, and the activity of inflammasome was reduced. Iraqi propolis-induced autophagy in in vitro and in vivo models decreased the levels of IL-1β and caspase-1. Results indicated that IL-1β pathway production is regulated by autophagy via two different novel mechanisms, namely, regulation of the activation of NLRC4 inflammasome and IL-1β targeting for lysosomal degradation.
Collapse
|
49
|
Yuan L, Liu J, Deng H, Gao C. Benzo[a]pyrene Induces Autophagic and Pyroptotic Death Simultaneously in HL-7702 Human Normal Liver Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:9763-9773. [PMID: 28990778 DOI: 10.1021/acs.jafc.7b03248] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
As a common polycyclic aromatic hydrocarbon compound, benzo[a]pyrene (BaP) is readily produced in processing of oil and fatty foods. It is not only a strong carcinogen but also a substance with strong immunotoxicity and reproduction toxicity. Autophagy and pyroptosis are two types of programmed cell death. Whether or not BaP damages body tissues via autophagy or pyroptosis remains unknown. The present study investigated the effects of BaP on autophagy and pyroptosis in HL-7702 cells. The results showed that BaP induced cell death in HL-7702 cells enhanced the intracellular levels of ROS and arrested the cell cycle at the S phase. Additionally, BaP resulted in cell death through autophagy and pyroptosis. Compared with the BaP group, the autophagy inhibitor 3-MA significantly (p < 0.01) inhibited the release of LDH by 70.53% ± 0.46 and NO by 50.36% ± 0.80, the increase of electrical conductivity by 12.08% ± 0.55, and the expressions of pyroptotic marker proteins (Caspase-1, Cox-2, IL-1β, IL-18). The pyroptosis inhibitor Ac-YVAD-CM also notably (p < 0.01) blocked BaP-induced autophagic cell death characterized by the increase of autophagic vacuoles and overexpression of Beclin-1 and LC3-II. In conclusion, BaP led to injury by inducing autophagy and pyroptosis simultaneously, the two of which coexisted and promoted each other in HL-7702 cells.
Collapse
Affiliation(s)
- Li Yuan
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University , Xi'an, 710119, China
| | - Junyi Liu
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University , Xi'an, 710119, China
| | - Hong Deng
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University , Xi'an, 710119, China
| | - Chunxia Gao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University , Xi'an, 710119, China
| |
Collapse
|
50
|
Bah A, Vergne I. Macrophage Autophagy and Bacterial Infections. Front Immunol 2017; 8:1483. [PMID: 29163544 PMCID: PMC5681717 DOI: 10.3389/fimmu.2017.01483] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/23/2017] [Indexed: 12/25/2022] Open
Abstract
Autophagy is a well-conserved lysosomal degradation pathway that plays key roles in bacterial infections. One of the most studied is probably xenophagy, the selective capture and degradation of intracellular bacteria by lysosomes. However, the impact of autophagy goes beyond xenophagy and involves intensive cross-talks with other host defense mechanisms. In addition, autophagy machinery can have non-canonical functions such as LC3-associated phagocytosis. In this review, we intend to summarize the current knowledge on the many functions of autophagy proteins in cell defenses with a focus on bacteria–macrophage interaction. We also present the strategies developed by pathogens to evade or to exploit this machinery in order to establish a successful infection. Finally, we discuss the opportunities and challenges of autophagy manipulation in improving therapeutics and vaccines against bacterial pathogens.
Collapse
Affiliation(s)
- Aïcha Bah
- Institut de Pharmacologie et de Biologie Structurale, UMR 5089 CNRS-Université de Toulouse, Toulouse, France
| | - Isabelle Vergne
- Institut de Pharmacologie et de Biologie Structurale, UMR 5089 CNRS-Université de Toulouse, Toulouse, France
| |
Collapse
|