1
|
Wei C, Xu Y, Zheng Y, Hong L, Lyu C, Li H, Cao B. The LTB4-BLT1 axis attenuates influenza-induced lung inflammation by suppressing NLRP3 activation. Cell Death Discov 2025; 11:148. [PMID: 40189592 PMCID: PMC11973165 DOI: 10.1038/s41420-025-02450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
The mortality associated with influenza A virus (IAV) infection typically results from excessive immune responses, leading to immunopathological lung damage and compromised pulmonary function. Various immunomodulators are seen beneficial when used in conjunction with direct anti-infection treatment. Leukotriene B4 (LTB4) is a derivative of arachidonic acid (AA) and has been shown to be advantageous for numerous infectious diseases, allergies, and autoimmune disorders. Nonetheless, the function of LTB4 in influenza infection remains unclear. This study demonstrates that LTB4 and its primary receptor BLT1, as opposed to the secondary receptor BLT2, act as a protective immune modulator during influenza infection in bone marrow-derived macrophages and mouse models. Mechanistically, LTB4 promotes K27-linked and K48-linked polyubiquitination of the NLRP3 protein at its K886 and K1023 sites via a cAMP/PKA-dependent pathway, which inhibits NLRP3 inflammasome assembly and thereby diminishes subsequent NLRP3 inflammasome activation. The consequent decline in the release of IL-1β and IL-18 leads to a reduction in inflammation caused by viral infection. Furthermore, the administration of a LTB4 treatment in a fatal IAV infection model can mitigate the excessive NLRP3 inflammasome activation and reduce IAV-induced severe pulmonary damage. These findings illustrate the protective function of LTB4 in fatal IAV infection by mitigating the severe inflammation induced by the virus.
Collapse
Affiliation(s)
- Cheng Wei
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yitian Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Zheng
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Lizhe Hong
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Lyu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haibo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Bin Cao
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
- New Cornerstone Science Laboratory, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
2
|
Balde A, Benjakul S, Nazeer RA. A review on NLRP3 inflammasome modulation by animal venom proteins/peptides: mechanisms and therapeutic insights. Inflammopharmacology 2025; 33:1013-1031. [PMID: 39934538 DOI: 10.1007/s10787-025-01656-7] [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: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025]
Abstract
The venom peptides from terrestrial as well as aquatic species have demonstrated potential in regulating the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a sophisticated assemblage present in immune cells responsible for detecting and responding to external mediators. The NLRP3 inflammasome plays a role in several pathological conditions such as type 2 diabetes, hyperglycemia, Alzheimer's disease, obesity, autoimmune disorders, and cardiovascular disorders. Venom peptides derived from animal venoms have been discovered to selectively induce certain signalling pathways, such as the NLRP3 inflammasome, mitogen-activated protein kinase (MAPK), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Experimental evidence has demonstrated that venom peptides can regulate the expression and activation of the NLRP3 inflammasome, resulting in the secretion of pro-inflammatory cytokines including interleukin (IL)-1β and IL-18. Furthermore, these peptides have been discovered to impede the activation of the NLRP3 inflammasome, therefore diminishing inflammation and tissue injury. The functional properties of venom proteins and peptides obtained from snakes, bees, wasps, and scorpions have been thoroughly investigated, specifically targeting the NLRP3 inflammasome pathway, venom proteins and peptides have shown promise as therapeutic agents for the treatment of certain inflammatory disorders. This review discusses the pathophysiology of NLRP3 inflammasome in the onset of various diseases, role of venom as therapeutics. Further, various venom components and their role in the modulation of NLRP3 inflammasome are discoursed. A substantial number of venomous animals and their toxins are yet unexplored, and to comprehensively grasp the mechanisms of action of them and their potential as therapeutic agents, additional research is required which can lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro Industry, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
- Department of Food and Nutrition, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India.
| |
Collapse
|
3
|
Sales LS, Silva-Sousa AC, Nascimento GC, Bel ED, Paula-Silva FWG. Effects of cannabidiol on biomineralization and inflammatory mediators expression in immortalized murine dental pulp cells and macrophages under pro-inflammatory conditions. J Dent 2025; 153:105535. [PMID: 39706322 DOI: 10.1016/j.jdent.2024.105535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
OBJECTIVES This study investigated the in vitro effects of cannabidiol (CBD) on dental pulp cells and macrophages under pro-inflammatory conditions. MATERIALS AND METHODS Mouse dental pulp cells (OD-21) were pre-stimulated with tumor necrosis factor alpha (10 ng/mL) or left untreated, then exposed to CBD at concentrations of 0.01 µM, 0.1 µM, 1 µM, and 10 µM for 24 h and 7 days. Cell viability was assessed using the MTT assay, while gene expression related to mineralization-Dentin Sialophosphoprotein (Dspp), Dentin Matrix Protein 1 (Dmp1), Runt-related transcription factor 2 (Runx2), TNF-α (Tnf), and prostaglandin-endoperoxide synthase 2 (Ptgs2) were analyzed via quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Mineralization nodule formation was evaluated using alizarin red staining. Macrophages (RAW 264.7) were stimulated with lipopolysaccharide (LPS) for 2 h before exposure to the same CBD concentrations. Data analysis included the Shapiro-Wilk normality test and comparisons using ANOVA and Tukey's post-hoc test (α = 0.05). RESULTS The findings indicated that CBD did not significantly affect OD-21 cell viability, except for the 10 µM concentration after 7 days (p < 0.05). CBD treatment promoted mineralization, with significant differences observed among groups (p < 0.05). Notably, Ptgs2 expression varied between time points, while Runx2 expression was significantly reduced at 24 h (p < 0.05). In macrophages, Ptgs2 and Tnf levels were downregulated by all tested CBD concentrations (p < 0.05). CONCLUSION These results indicate that cannabidiol positively influence the biomineralization process and modulate inflammatory mediator expression. CLINICAL RELEVANCE Our research indicates that cannabidiol presents biomineralization potential within inflammatory contexts, implying its potential as a promisor bioactive substance for regenerating oral tissues by interacting with cells and tissues to induce specific responses.
Collapse
Affiliation(s)
- Larissa Sthefani Sales
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Alice Correa Silva-Sousa
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Elaine Del Bel
- Department of Basic and Oral Biology, University of São Paulo, Ribeirão Preto, SP, São Paulo, Brazil
| | | |
Collapse
|
4
|
Reis MB, Arantes EC. Immunosuppressive therapies in scorpion envenomation: new perspectives for treatment. FRONTIERS IN TOXICOLOGY 2024; 6:1503055. [PMID: 39628768 PMCID: PMC11611809 DOI: 10.3389/ftox.2024.1503055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/06/2024] [Indexed: 12/06/2024] Open
Abstract
Scorpion envenoming is a relevant and neglected public health problem in some countries. The use of antivenom is widespread in many regions, targeting specific species of scorpions. However, the uncontrolled proliferation and adaptation of these animals to urban environments, combined with limited access to treatments in remote areas and delays in antivenom administration contribute to a significant number of fatalities from scorpion-related incidents. In recent decades, new research has revealed that the immune system plays an important role in triggering immunopathological reactions during scorpion envenoming, which places it as a therapeutic target; however, few clinical studies have been conducted. This work provides a review of the main immunopathological aspects of scorpion envenoming, as well as the clinical trials conducted to date on the use of corticosteroids for the treatment of scorpionism. We highlight emerging treatment perspectives as well as the need for further clinical trials. The use of corticosteroids in scorpionism, when appropriate, could significantly enhance access to treatment and help reduce fatalities associated with scorpion stings.
Collapse
Affiliation(s)
- Mouzarllem Barros Reis
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | | |
Collapse
|
5
|
Issy AC, Pedrazzi JF, Nascimento GC, Faccioli LH, Del Bel E. Impact of 5-Lipoxygenase Deficiency on Dopamine-Mediated Behavioral Responses. Neurotox Res 2024; 42:42. [PMID: 39365372 DOI: 10.1007/s12640-024-00720-4] [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: 03/04/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The 5-lipoxygenase/leukotriene system has been implicated in both physiological and pathological states within the central nervous system. Understanding how this system interacts with the dopaminergic system could provide valuable insights into dopamine-related pathologies. This study focused on examining both motor and non-motor dopamine-related responses in 5-lipoxygenase/leukotriene-deficient mice. We used pharmacological agents such as amphetamine, apomorphine, and reserpine to challenge the dopaminergic system, evaluating their effects on prepulse inhibition reaction (PPI), general motor activity, and oral involuntary movements. Additionally, we analyzed striatal glial marker expression (GFAP and Iba-1) in reserpine-treated mice. The 5-lipoxygenase/leukotriene-deficient mice exhibited increased spontaneous locomotor activity, including both horizontal and vertical exploration, along with stereotyped behavior compared to wild-type mice. This hyperactivity was reduced by acute apomorphine treatment. Although basal PPI responses were unchanged, 5-lipoxygenase/leukotriene-deficient mice displayed a significant reduction in susceptibility to amphetamine-induced PPI disruption. Conversely, these mice were more vulnerable to reserpine-induced involuntary movements. There were no significant differences in the basal expression of striatal GFAP and Iba-1 positive cells between 5-lipoxygenase/leukotriene-deficient and wild-type mice. However, reserpine treatment significantly increased GFAP immunoreactivity in wild-type mice, an effect not observed in 5-lipoxygenase-deficient mice. Additionally, the percentage of activated microglia was significantly higher in reserpine-treated wild-type mice, an effect absents in 5-lipoxygenase/leukotriene-deficient mice. Our findings suggest that 5-lipoxygenase/leukotriene deficiency leads to a distinctive dopaminergic phenotype, indicating that leukotrienes may influence the modulation of dopamine-mediated responses.
Collapse
Affiliation(s)
- Ana Carolina Issy
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - João Francisco Pedrazzi
- Department of Neuroscience and Behavior Sciences, Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Glauce Crivelaro Nascimento
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Lúcia Helena Faccioli
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Elaine Del Bel
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
6
|
Sun J, Liu J, Liu M, Bi X, Huang C. New perspective for pathomechanism and clinical applications of animal toxins: Programmed cell death. Toxicon 2024; 249:108071. [PMID: 39134227 DOI: 10.1016/j.toxicon.2024.108071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
Various animal toxins pose a significant threat to human safety, necessitating urgent attention to their treatment and research. The clinical potential of programmed cell death (PCD) is widely regarded as a target for envenomation, given its crucial role in regulating physiological and pathophysiological processes. Current research on animal toxins examines their specific components in pathomechanisms and injuries, as well as their clinical applications. This review explores the relationship between various toxins and several types of PCD, such as apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis, to provide a reference for future understanding of the pathophysiology of toxins and the development of their potential clinical value.
Collapse
Affiliation(s)
- Jiaqi Sun
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jiahao Liu
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Meiling Liu
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xiaowen Bi
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Chunhong Huang
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
7
|
Brady A, Mora Martinez LC, Hammond B, Whitefoot-Keliin KM, Haribabu B, Uriarte SM, Lawrenz MB. Distinct mechanisms of type 3 secretion system recognition control LTB4 synthesis in neutrophils and macrophages. PLoS Pathog 2024; 20:e1012651. [PMID: 39423229 PMCID: PMC11524448 DOI: 10.1371/journal.ppat.1012651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/30/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Leukotriene B4 (LTB4) is an inflammatory lipid produced in response to pathogens that is critical for initiating the inflammatory cascade needed to control infection. However, during plague, Yersinia pestis inhibits the timely synthesis of LTB4 and subsequent inflammation. Using bacterial mutants, we previously determined that Y. pestis inhibits LTB4 synthesis via the action of the Yop effector proteins that are directly secreted into host cells through a type 3 secretion system (T3SS). Here, we show that the T3SS is the primary pathogen associated molecular pattern (PAMP) required for production of LTB4 in response to both Yersinia and Salmonella. However, we also unexpectantly discovered that T3SS-mediated LTB4 synthesis by neutrophils and macrophages require the activation of two distinctly different host signaling pathways. We identified that phagocytosis and the NLRP3/CASP1 inflammasome significantly impact LTB4 synthesis by macrophages but not neutrophils. Instead, the SKAP2/PLC signaling pathway is required for T3SS-mediated LTB4 production by neutrophils. Finally, while recognition of the T3SS is required for LTB4 production, we also discovered that a second unrelated PAMP-mediated signal activates the MAP kinase pathway needed for synthesis. Together, these data demonstrate significant differences in the host factors and signaling pathways required by macrophages and neutrophils to quickly produce LTB4 in response to bacteria. Moreover, while macrophages and neutrophils might rely on different signaling pathways for T3SS-dependent LTB4 synthesis, Y. pestis has evolved virulence mechanisms to counteract this response by either leukocyte to inhibit LTB4 synthesis and colonize the host.
Collapse
Affiliation(s)
- Amanda Brady
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Leonardo C. Mora Martinez
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Benjamin Hammond
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Kaitlyn M. Whitefoot-Keliin
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Center for Microbiomics, Inflammation and Pathogenicity, Louisville, Kentucky, United States of America
| | - Silvia M. Uriarte
- Deptartment of Oral Immunology & Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| | - Matthew B. Lawrenz
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| |
Collapse
|
8
|
Atalay Ekiner S, Gęgotek A, Skrzydlewska E. Inflammasome activity regulation by PUFA metabolites. Front Immunol 2024; 15:1452749. [PMID: 39290706 PMCID: PMC11405227 DOI: 10.3389/fimmu.2024.1452749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Oxidative stress and the accompanying chronic inflammation constitute an important metabolic problem that may lead to pathology, especially when the body is exposed to physicochemical and biological factors, including UV radiation, pathogens, drugs, as well as endogenous metabolic disorders. The cellular response is associated, among others, with changes in lipid metabolism, mainly due to the oxidation and the action of lipolytic enzymes. Products of oxidative fragmentation/cyclization of polyunsaturated fatty acids (PUFAs) [4-HNE, MDA, 8-isoprostanes, neuroprostanes] and eicosanoids generated as a result of the enzymatic metabolism of PUFAs significantly modify cellular metabolism, including inflammation and the functioning of the immune system by interfering with intracellular molecular signaling. The key regulators of inflammation, the effectiveness of which can be regulated by interacting with the products of lipid metabolism under oxidative stress, are inflammasome complexes. An example is both negative or positive regulation of NLRP3 inflammasome activity by 4-HNE depending on the severity of oxidative stress. 4-HNE modifies NLRP3 activity by both direct interaction with NLRP3 and alteration of NF-κB signaling. Furthermore, prostaglandin E2 is known to be positively correlated with both NLRP3 and NLRC4 activity, while its potential interference with AIM2 or NLRP1 activity is unproven. Therefore, the influence of PUFA metabolites on the activity of well-characterized inflammasome complexes is reviewed.
Collapse
Affiliation(s)
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
9
|
Mabunda IG, Zinyemba NK, Pillay S, Offor BC, Muller B, Piater LA. The geographical distribution of scorpions, implication of venom toxins, envenomation, and potential therapeutics in Southern and Northern Africa. Toxicol Res (Camb) 2024; 13:tfae118. [PMID: 39100857 PMCID: PMC11298049 DOI: 10.1093/toxres/tfae118] [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: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024] Open
Abstract
Scorpions are predatory arachnids whose venomous sting primarily affects people in tropical and subtropical regions. Most scorpion stings can only cause localized pain without severe envenomation. Less than one-third of the stings cause systemic envenoming and possibly lead to death. About 350,000 scorpion stings in Northern Africa are recorded yearly, resulting in about 810 deaths. In Eastern/Southern Africa, there are about 79,000 stings recorded yearly, resulting in 245 deaths. Farmers and those living in poverty-stricken areas are among the most vulnerable to getting stung by scorpions. However, compared to adults, children are at greater risk of severe envenomation. Scorpion venom is made up of complex mixtures dominated by peptides and proteins that confer its potency and toxicity. These venom toxins have intra- and interspecies variations associated with the scorpion's habitat, sex, diet, and age. These variations alter the activity of antivenoms used to treat scorpion sting envenomation. Thus, the study of the proteome composition of medically important scorpion venoms needs to be scaled up along their geographical distribution and contributions to envenomation in Southern and Northern Africa. This will help the production of safer, more effective, and broad-spectrum antivenoms within these regions. Here, we review the clinical implications of scorpion sting envenomation in Southern and Northern Africa. We further highlight the compositions of scorpion venoms and tools used in scorpion venomics. We discuss current antivenoms used against scorpion sting envenomation and suggestions for future production of better antivenoms or alternatives. Finally, we discuss the therapeutic properties of scorpion venom.
Collapse
Affiliation(s)
- Isac G Mabunda
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Nodji K Zinyemba
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Shanelle Pillay
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Benedict C Offor
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Beric Muller
- South Africa Venom Suppliers cc, 41 Louis, Trichardt 0920, South Africa
| | - Lizelle A Piater
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| |
Collapse
|
10
|
Brady A, Mora-Martinez LC, Hammond B, Haribabu B, Uriarte SM, Lawrenz MB. Distinct Mechanisms of Type 3 Secretion System Recognition Control LTB 4 Synthesis in Neutrophils versus Macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601466. [PMID: 39005373 PMCID: PMC11244889 DOI: 10.1101/2024.07.01.601466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Leukotriene B4 (LTB4) is critical for initiating the inflammatory cascade in response to infection. However, Yersinia pestis colonizes the host by inhibiting the timely synthesis of LTB4 and inflammation. Here, we show that the bacterial type 3 secretion system (T3SS) is the primary pathogen associated molecular pattern (PAMP) responsible for LTB4 production by leukocytes in response to Yersinia and Salmonella, but synthesis is inhibited by the Yop effectors during Yersinia interactions. Moreover, we unexpectedly discovered that T3SS-mediated LTB4 synthesis by neutrophils and macrophages require two distinct host signaling pathways. We show that the SKAP2/PLC signaling pathway is essential for LTB4 production by neutrophils but not macrophages. Instead, phagocytosis and the NLRP3/CASP1 inflammasome are needed for LTB4 synthesis by macrophages. Finally, while recognition of the T3SS is required for LTB4 production, we also discovered a second unrelated PAMP-mediated signal independently activates the MAP kinase pathway needed for LTB4 synthesis. Together, these data demonstrate significant differences in the signaling pathways required by macrophages and neutrophils to quickly respond to bacterial infections.
Collapse
Affiliation(s)
- Amanda Brady
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Leonardo C. Mora-Martinez
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Benjamin Hammond
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Center for Microbiomics, Inflammation and Pathogenicity, Louisville, Kentucky, United States of America
| | - Silvia M. Uriarte
- Deptartment of Oral Immunology & Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| | - Matthew B. Lawrenz
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| |
Collapse
|
11
|
Gunas V, Maievskyi O, Synelnyk T, Raksha N, Vovk T, Halenova T, Savchuk O, Gunas I. Cytokines and their regulators in rat lung following scorpion envenomation. Toxicon X 2024; 22:100198. [PMID: 38633505 PMCID: PMC11022085 DOI: 10.1016/j.toxcx.2024.100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
Nowadays, more than two billion inhabitants of underdeveloped tropical and subtropical countries are at risk of being stung by scorpions. Scorpion stings annually cause 2000-3000 deaths as they can lead to the respiratory and/or cardiovascular complications. Pathogenesis of lung damage under scorpion envenomation is often comprehensive. Respiratory failure can have a cardiogenic origin, associated with venom neurotoxin action. However, some venom components can stimulate pro-inflammatory signaling cascades followed by cytokines synthesis, recruit and activate immune cells, participating in the inflammatory response in lung injury. Scorpions of the Leiurus genus ("deathstalker") are one of the most dangerous Arthropoda. To date, 22 species of this genus have been described, but the venom composition and the mechanisms of tissues damage under envenomation have been studied to some extent only for L. quinquestriatus, L. hebraeus, and L. abdullahbayrami. Scorpions of L. macroctenus species are expected to be very hazardous, but the possibility of their venom cause inflammation in the lung tissue has not been investigated to date. Therefore, in this study, we focused on evaluating the levels of cytokines and their regulators - transcription factors (HIF-1α and NF-κB) and growth factors (FGF-2, VEGF, and EGF) - in rat lung homogenates after L. macroctenus envenomation. The results revealed a decrease in the levels of most pro-inflammatory cytokines (IL-6, IL-8, IL-1β and TNF-α) with simultaneous rise in the content of both anti-inflammatory cytokines (IL-4 and IL-10) and interferon-γ. Furthermore, the levels of all researched transcription factors and growth factors were shown to be increased too. The detected changes peak occurred at 24 h, whereas a tendency towards all indicators values normalization was observed in 72 h after venom injection. Thus, our results did not reveal signs of a classic inflammatory process in the lungs of rats injected with L. macroctenus venom. However, the obtained data indicate venom influence both on cytokine profile and on their regulators content in the rat lungs, which is a feature of certain alterations in the innate immune response, caused by studied venom components. But, the mechanisms of the changes we found require additional researches.
Collapse
Affiliation(s)
- Valery Gunas
- Department of Forensic Medicine and Law, National Pirogov Memorial Medical University, Vinnytsya, Ukraine
| | - Oleksandr Maievskyi
- Department of Clinical Medicine, Educational and Scientific Center Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tatyana Synelnyk
- Department of Biochemistry, Educational and Scientific Center Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Nataliia Raksha
- Department of Biochemistry, Educational and Scientific Center Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetiana Vovk
- Department of Biochemistry, Educational and Scientific Center Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetiana Halenova
- Department of Biochemistry, Educational and Scientific Center Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olexiy Savchuk
- Department of Biochemistry, Educational and Scientific Center Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Igor Gunas
- Department of Human Anatomy, National Pirogov Memorial Medical University, Vinnytsya, Ukraine
| |
Collapse
|
12
|
Nandana MB, Bharatha M, Praveen R, Nayaka S, Vishwanath BS, Rajaiah R. Dimethyl ester of bilirubin ameliorates Naja naja snake venom-induced lung toxicity in mice via inhibiting NLRP3 inflammasome and MAPKs activation. Toxicon 2024; 244:107757. [PMID: 38740099 DOI: 10.1016/j.toxicon.2024.107757] [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: 04/08/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Naja naja snake bite causes thousands of deaths worldwide in a year. N. naja envenomed victims exhibit both local and systemic reactions that potentially lead to death. In clinical practice, pulmonary complications in N. naja envenomation are commonly encountered. However, the molecular mechanisms underlying N. naja venom-induced lung toxicity remain unknown. Here, we reasoned that N. naja venom-induced lung toxicity is prompted by NLRP3 inflammasome and MAPKs activation in mice. Treatment with dimethyl ester of bilirubin (BD1), significantly inhibited the N. naja venom-induced activation of NLRP3 inflammasome and MAPKs both in vivo and in vitro (p < 0.05). Further, BD1 reduced N. naja venom-induced recruitment of inflammatory cells, and hemorrhage in the lung toxicity examined by histopathology. BD1 also diminished N. naja venom-induced local toxicities in paw edema and myotoxicity in mice. Furthermore, BD1 was able to enhance the survival time against N. naja venom-induced mortality in mice. In conclusion, the present data showed that BD1 alleviated N. naja venom-induced lung toxicity by inhibiting NLRP3 inflammasome and MAPKs activation. Small molecule inhibitors that intervene in venom-induced toxicities may have therapeutic applications complementing anti-snake venom.
Collapse
Affiliation(s)
- Manuganahalli B Nandana
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India; Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India
| | - Madeva Bharatha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India
| | - Raju Praveen
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India
| | - Spandan Nayaka
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India.
| | - Rajesh Rajaiah
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India.
| |
Collapse
|
13
|
Wiezel GA, Oliveira IS, Reis MB, Ferreira IG, Cordeiro KR, Bordon KCF, Arantes EC. The complex repertoire of Tityus spp. venoms: Advances on their composition and pharmacological potential of their toxins. Biochimie 2024; 220:144-166. [PMID: 38176606 DOI: 10.1016/j.biochi.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/30/2023] [Accepted: 12/30/2023] [Indexed: 01/06/2024]
Abstract
Animal venoms are a rich and complex source of components, including peptides (such as neurotoxins, anionic peptides and hypotensins), lipids, proteins (such as proteases, hyaluronidases and phospholipases) and inorganic compounds, which affect all biological systems of the envenoming victim. Their action may result in a wide range of clinical manifestations, including tachy/bradycardia, hyper/hypotension, disorders in blood coagulation, pain, edema, inflammation, fever, muscle paralysis, coma and even death. Scorpions are one of the most studied venomous animals in the world and interesting bioactive molecules have been isolated and identified from their venoms over the years. Tityus spp. are among the scorpions with high number of accidents reported in the Americas, especially in Brazil. Their venoms have demonstrated interesting results in the search for novel agents with antimicrobial, anti-viral, anti-parasitic, hypotensive, immunomodulation, anti-insect, antitumor and/or antinociceptive activities. Furthermore, other recent activities still under investigation include drug delivery action, design of anti-epileptic drugs, investigation of sodium channel function, treatment of erectile disfunction and priapism, improvement of scorpion antivenom and chelating molecules activity. In this scenario, this paper focuses on reviewing advances on Tityus venom components mainly through the modern omics technologies as well as addressing potential therapeutic agents from their venoms and highlighting this abundant source of pharmacologically active molecules with biotechnological application.
Collapse
Affiliation(s)
- Gisele A Wiezel
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida Do Café s/n, Ribeirão Preto, SP, Brazil.
| | - Isadora S Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida Do Café s/n, Ribeirão Preto, SP, Brazil; Department of Biotechnology and Biomedicine, Technical University of Denmark, Søtolfts Plads, Building 239 Room 006, Kongens Lyngby, 2800, Denmark.
| | - Mouzarllem B Reis
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida Do Café s/n, Ribeirão Preto, SP, Brazil.
| | - Isabela G Ferreira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida Do Café s/n, Ribeirão Preto, SP, Brazil.
| | - Kalynka R Cordeiro
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida Do Café s/n, Ribeirão Preto, SP, Brazil.
| | - Karla C F Bordon
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida Do Café s/n, Ribeirão Preto, SP, Brazil.
| | - Eliane C Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida Do Café s/n, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
14
|
Pereira M, Liang J, Edwards-Hicks J, Meadows AM, Hinz C, Liggi S, Hepprich M, Mudry JM, Han K, Griffin JL, Fraser I, Sack MN, Hess C, Bryant CE. Arachidonic acid inhibition of the NLRP3 inflammasome is a mechanism to explain the anti-inflammatory effects of fasting. Cell Rep 2024; 43:113700. [PMID: 38265935 PMCID: PMC10940735 DOI: 10.1016/j.celrep.2024.113700] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/27/2023] [Accepted: 01/05/2024] [Indexed: 01/26/2024] Open
Abstract
Elevated interleukin (IL)-1β levels, NLRP3 inflammasome activity, and systemic inflammation are hallmarks of chronic metabolic inflammatory syndromes, but the mechanistic basis for this is unclear. Here, we show that levels of plasma IL-1β are lower in fasting compared to fed subjects, while the lipid arachidonic acid (AA) is elevated. Lipid profiling of NLRP3-stimulated mouse macrophages shows enhanced AA production and an NLRP3-dependent eicosanoid signature. Inhibition of cyclooxygenase by nonsteroidal anti-inflammatory drugs decreases eicosanoid, but not AA, production. It also reduces both IL-1β and IL-18 production in response to NLRP3 activation. AA inhibits NLRP3 inflammasome activity in human and mouse macrophages. Mechanistically, AA inhibits phospholipase C activity to reduce JNK1 stimulation and hence NLRP3 activity. These data show that AA is an important physiological regulator of the NLRP3 inflammasome and explains why fasting reduces systemic inflammation and also suggests a mechanism to explain how nonsteroidal anti-inflammatory drugs work.
Collapse
Affiliation(s)
- Milton Pereira
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jonathan Liang
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK; Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Joy Edwards-Hicks
- The Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Cambridge, UK
| | - Allison M Meadows
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, MD, USA; Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Christine Hinz
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Sonia Liggi
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Julian L Griffin
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Iain Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Christoph Hess
- The Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Cambridge, UK
| | - Clare E Bryant
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
15
|
Nandana MB, Bharatha M, Vishwanath BS, Rajaiah R. Naja naja snake venom-induced local toxicities in mice is by inflammasome activation. Toxicon 2024; 238:107590. [PMID: 38163462 DOI: 10.1016/j.toxicon.2023.107590] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
Snake bite envenomation causes tissue damage resulting in acute and chronic inflammatory responses. Inflammasome activation is one of the factors involved in tissue damage in a mouse model of snake envenomation. The present study examines the potency of Indian Big Four snake venoms in the activation of inflammasome and its role in local and systemic tissue toxicity. Among Indian Big Four snake venoms, Naja naja venom activated NLRP3 inflammasome in mouse macrophages. Activation of NLRP3 inflammasome was also observed in mouse foot paw and thigh muscle upon administration of N. naja venom. Intraperitoneal administration of N. naja venom cause systemic lung damage showed activation of NLRP3 inflammasome. Treatment with MCC950, a selective NLRP3 inflammasome inhibitor effectively inhibited N. naja venom-induced activation of caspase-1 and liberation of IL-1β in macrophages. In mice, MCC950 partially inhibited the activation of NLRP3 inflammasome in N. naja venom administered foot paw and thigh muscle. In conclusion, the present data showed that inflammasome is one of the host responses involved in N. naja snake venom-induced toxicities. The inhibition of inflammasome activation will provide new insight into better management of snake bite-induced local tissue damage.
Collapse
Affiliation(s)
- Manuganahalli B Nandana
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India; Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India
| | - Madeva Bharatha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India.
| | - Rajesh Rajaiah
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, Karnataka, 570006, India.
| |
Collapse
|
16
|
Matkivska R, Samborska I, Maievskyi O. Effect of animal venom toxins on the main links of the homeostasis of mammals (Review). Biomed Rep 2024; 20:16. [PMID: 38144889 PMCID: PMC10739175 DOI: 10.3892/br.2023.1704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/15/2023] [Indexed: 12/26/2023] Open
Abstract
The human body is affected by environmental factors. The dynamic balance between the organism and its environment results from the influence of natural, anthropogenic and social aspects. The factors of exogenous origin determine development of adaptive changes. The present article summarises the mechanisms of animal venom toxins and homeostasis disruption in the body of mammals. The mechanisms underlying pathological changes are associated with shifts in biochemical reactions. Components of the immune, nervous and endocrine systems are key in the host defense and adaptation processes in response to venom by triggering signalling pathways (PI3kinase pathway, arachidonic acid cascade). Animal venom toxins initiate the development of inflammatory processes, the synthesis of pro-inflammatory mediators (cytokines), ROS, proteolytic enzymes, activate the migration of leukocytes and macrophages. Keratinocytes and endothelial cells act as protective barriers under the action of animal venom toxins on the body of mammals. In addition, the formation of pores in cell membranes, structural changes in cell ion channels are characteristic of the action of animal venom toxins.
Collapse
Affiliation(s)
- Ruzhena Matkivska
- Department of Descriptive and Clinical Anatomy, Bogomolets National Medical University, Kyiv 03680, Ukraine
| | - Inha Samborska
- Department of Biological and General Chemistry, National Pirogov Memorial Medical University, Vinnytsya 21018, Ukraine
| | - Oleksandr Maievskyi
- Department of Clinical Medicine, Educational and Scientific Center ‘Institute of Biology and Medicine’ of Taras Shevchenko National University of Kyiv, Kyiv 03127, Ukraine
| |
Collapse
|
17
|
Brady A, Sheneman KR, Pulsifer AR, Price SL, Garrison TM, Maddipati KR, Bodduluri SR, Pan J, Boyd NL, Zheng JJ, Rai SN, Hellmann J, Haribabu B, Uriarte SM, Lawrenz MB. Type 3 secretion system induced leukotriene B4 synthesis by leukocytes is actively inhibited by Yersinia pestis to evade early immune recognition. PLoS Pathog 2024; 20:e1011280. [PMID: 38271464 PMCID: PMC10846697 DOI: 10.1371/journal.ppat.1011280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 02/06/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Subverting the host immune response to inhibit inflammation is a key virulence strategy of Yersinia pestis. The inflammatory cascade is tightly controlled via the sequential action of lipid and protein mediators of inflammation. Because delayed inflammation is essential for Y. pestis to cause lethal infection, defining the Y. pestis mechanisms to manipulate the inflammatory cascade is necessary to understand this pathogen's virulence. While previous studies have established that Y. pestis actively inhibits the expression of host proteins that mediate inflammation, there is currently a gap in our understanding of the inflammatory lipid mediator response during plague. Here we used the murine model to define the kinetics of the synthesis of leukotriene B4 (LTB4), a pro-inflammatory lipid chemoattractant and immune cell activator, within the lungs during pneumonic plague. Furthermore, we demonstrated that exogenous administration of LTB4 prior to infection limited bacterial proliferation, suggesting that the absence of LTB4 synthesis during plague contributes to Y. pestis immune evasion. Using primary leukocytes from mice and humans further revealed that Y. pestis actively inhibits the synthesis of LTB4. Finally, using Y. pestis mutants in the Ysc type 3 secretion system (T3SS) and Yersinia outer protein (Yop) effectors, we demonstrate that leukocytes recognize the T3SS to initiate the rapid synthesis of LTB4. However, several Yop effectors secreted through the T3SS effectively inhibit this host response. Together, these data demonstrate that Y. pestis actively inhibits the synthesis of the inflammatory lipid LTB4 contributing to the delay in the inflammatory cascade required for rapid recruitment of leukocytes to sites of infection.
Collapse
Affiliation(s)
- Amanda Brady
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Katelyn R. Sheneman
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Amanda R. Pulsifer
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Sarah L. Price
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Taylor M. Garrison
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Krishna Rao Maddipati
- Department of Pathology, Lipidomics Core Facility, Wayne State University, Detroit, Michigan, United States of America
| | - Sobha R. Bodduluri
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Jianmin Pan
- Biostatistics and Bioinformatics Facility, Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Nolan L. Boyd
- Center for Cardiometabolic Science, Christina Lee Brown Environment Institute, Division of Environmental Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Jing-Juan Zheng
- Center for Cardiometabolic Science, Christina Lee Brown Environment Institute, Division of Environmental Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Shesh N. Rai
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Jason Hellmann
- Center for Cardiometabolic Science, Christina Lee Brown Environment Institute, Division of Environmental Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Silvia M. Uriarte
- Deptartment of Oral Immunology & Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
| | - Matthew B. Lawrenz
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| |
Collapse
|
18
|
Tao Q, Liu XW, Zhang ZD, Ma N, Lu XR, Ge WB, Li JY, Yang YJ. Protective Effect and Mechanism of Aspirin Eugenol Ester on Lipopolysaccharide-Induced Intestinal Barrier Injury. Int J Mol Sci 2023; 24:17434. [PMID: 38139262 PMCID: PMC10743450 DOI: 10.3390/ijms242417434] [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: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Intestinal inflammation is a complex and recurrent inflammatory disease. Pharmacological and pharmacodynamic experiments showed that aspirin eugenol ester (AEE) has good anti-inflammatory, antipyretic, and analgesic effects. However, the role of AEE in regulating intestinal inflammation has not been explored. This study aimed to investigate whether AEE could have a protective effect on LPS-induced intestinal inflammation and thus help to alleviate the damage to the intestinal barrier. This was assessed with an inflammation model in Caco-2 cells and in rats induced with LPS. The expression of inflammatory mediators, intestinal epithelial barrier-related proteins, and redox-related signals was analyzed using an enzyme-linked immunosorbent assay (ELISA), Western blotting, immunofluorescence staining, and RT-qPCR. Intestinal damage was assessed by histopathological examination. Changes in rat gut microbiota and their functions were detected by the gut microbial metagenome. AEE significantly reduced LPS-induced pro-inflammatory cytokine levels (p < 0.05) and oxidative stress levels in Caco-2 cells and rats. Compared with the LPS group, AEE could increase the relative expression of Occludin, Claudin-1, and zonula occludens-1 (ZO-1) and decrease the relative expression of kappa-B (NF-κB) and matrix metalloproteinase-9. AEE could significantly improve weight loss, diarrhea, reduced intestinal muscle thickness, and intestinal villi damage in rats. Metagenome results showed that AEE could regulate the homeostasis of the gut flora and alter the relative abundance of Firmicutes and Bacteroidetes. Flora enrichment analysis indicated that the regulation of gut flora with AEE may be related to the regulation of glucose metabolism and energy metabolism. AEE could have positive effects on intestinal inflammation-related diseases.
Collapse
Affiliation(s)
- Qi Tao
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Xi-Wang Liu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Zhen-Dong Zhang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Ning Ma
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China;
| | - Xiao-Rong Lu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Wen-Bo Ge
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Jian-Yong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Ya-Jun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| |
Collapse
|
19
|
Nascimento MT, Viana DL, Peixoto FC, Arruda SM, Carvalho EM, Carvalho LP. Prostaglandin E2 contributes to L. braziliensis survival and therapeutic failure in cutaneous leishmaniasis. Emerg Microbes Infect 2023; 12:2261565. [PMID: 37729084 PMCID: PMC10540647 DOI: 10.1080/22221751.2023.2261565] [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: 06/27/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023]
Abstract
Patients with cutaneous leishmaniasis (CL) present an exacerbated inflammatory response associated with tissue damage and ulcer development. In recent years, higher rates of failure to pentavalent antimoniate therapy have been observed, yet the underlying reason remains poorly understood. We hypothesize that the eicosanoid PGE2 favours the establishment of infection by L. braziliensis, which contributes to therapeutic failure. The aim of the present study was to investigate the influence of PGE2 on the survival of L. braziliensis in macrophages and rates of therapeutic failure in CL patients. PGE2, an eicosanoid derived from the metabolism of arachidonic acid by the COX-2 enzyme, plays several roles in immune response. We found that increased PGE2 decreases the microbicidal function of macrophages and is associated with disease severity and therapeutic failure. Additionally, the neutralization of COX-2 by NS398, a selective NSAID, increases the ability of macrophages to kill L. braziliensis and protects against the pathological inflammatory response. Our data suggest that NS398 may serve as an adjunct treatment for CL patients.
Collapse
Affiliation(s)
- Maurício T. Nascimento
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Serviço de Imunologia, SIM, Complexo Universitário Professor Edgar Santos, COM-HUPES, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
| | - Débora L. Viana
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
| | - Fábio C. Peixoto
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
| | - Sérgio M. Arruda
- Laboratório Avançado de Saúde Pública, LASP, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
| | - Edgar M. Carvalho
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Serviço de Imunologia, SIM, Complexo Universitário Professor Edgar Santos, COM-HUPES, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
- Instituto de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, Brazil
| | - Lucas P. Carvalho
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Serviço de Imunologia, SIM, Complexo Universitário Professor Edgar Santos, COM-HUPES, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
- Instituto de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, Brazil
| |
Collapse
|
20
|
Reis MB, Ramos SG, Rocha LB, Faccioli LH. Mitochondrial swelling in cardiomyocytes: Insights from a murine model of Tityus serrulatus scorpion envenomation. Toxicon 2023; 233:107259. [PMID: 37595687 DOI: 10.1016/j.toxicon.2023.107259] [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: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Immune system hyperactivation is involved with clinical severity and pathological alterations during scorpion envenomation. In a murine model, mice inoculated with a lethal dose of Tityus serrulatus scorpion venom presented mitochondrial swelling in cardiomyocytes, with other structures such as sarcomeres and intercalated disks preserved. Treatment with dexamethasone or knockout animals to the interleukin-1β receptor do not undergo mitochondrial changes in cardiomyocytes during envenomation.
Collapse
Affiliation(s)
- Mouzarllem Barros Reis
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Faculdade de Medicina, Centro Universitário Barão de Mauá, Ribeirão Preto, São Paulo, Brazil; Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Simone Gusmão Ramos
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil
| | - Lenaldo Branco Rocha
- Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Minas Gerais, Brazil
| | - Lúcia Helena Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
21
|
Reis MB, Martins JG, Bordon KDCF, de Campos Fraga-Silva TF, de Lima Procópio RE, de Almeida BRR, Bonato VLD, Arantes EC. Pioneering in vitro characterization of macrophage response induced by scorpion venoms from the Brazilian Amazon. Toxicon 2023; 230:107171. [PMID: 37211059 DOI: 10.1016/j.toxicon.2023.107171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/07/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
There are several scorpion species of medical relevance around the world. Some of them are well characterized by their toxins and clinical outcomes. Brazilian Amazon has a great amount of these arthropods that have an impact in the scorpionism events specifically in this region of Brazil. Recently, several studies pointed out the immune system activation during scorpion envenouming as an important facet of scorpionism, inducing a sepsis-like state that culminates in clinical severity and death. In this work, we characterized the macrophage response of three species of clinical relevance in Brazilian Amazon: Tityus silvestris, T. metuendus and T. obscurus and one specie with no toxic effects to humans, Brotheas amazonicus. All the four species analyzed were able to induce pro- and anti-inflammatory cytokine production in a J774.1 murine macrophage model. This activation was dependent on TLR2/TLR4/MyD88 activation and abolished by TLRs antagonists. These results suggest that the venoms of the four species analyzed were able to induce macrophage response in agreement to the well-established immune activation by T. serrulatus venom. Our findings provide new insights into the clinical repercussions of scorpionism of uncharacterized species and point to new biotechnological applications of these venoms and possible supportive therapies in scorpionism.
Collapse
Affiliation(s)
- Mouzarllem Barros Reis
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil; School of Medicine, Centro Universitário Barão de Mauá (CBM), Ribeirão Preto, SP, Brazil
| | - Jonas Gama Martins
- Graduate Program in Genetics, Conservation and Evolutionary Biology (PPG GCBEv), National Institute for Amazon Research (INPA), Manaus, AM, Brazil
| | - Karla de Castro Figueiredo Bordon
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Thais Fernanda de Campos Fraga-Silva
- Program of Basic and Applied Immunology, And Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Rudi Emerson de Lima Procópio
- Graduate Program in Biotechnology and Natural Resources of Amazon, University of the State of Amazonas (UEA), Manaus, AM, Brazil
| | | | - Vânia Luiza Deperon Bonato
- Program of Basic and Applied Immunology, And Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Eliane Candiani Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
22
|
Acunha T, Rocha BA, Nardini V, Barbosa F, Faccioli LH. Lipidomic profiling of the Brazilian yellow scorpion venom: new insights into inflammatory responses following Tityus serrulatus envenomation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2023; 86:283-295. [PMID: 36895096 DOI: 10.1080/15287394.2023.2188896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Due to the high prevalence and clinical relevance, scorpionism is a critical public health issue in several Brazilian regions. Tityus serrulatus, commonly known as the Brazilian yellow scorpion, is the most venomous genus found in Brazilian fauna and associated with severe clinical manifestations such as localized pain, hypertension, sweating, tachycardia and complex hyperinflammatory responses. In general, T. serrulatus venom contains a complex mixture of active compounds, including proteins, peptides, and amino acids. Although knowledge of the protein fractions of scorpion venom is available, venom lipid components are not yet comprehensively known. The aim of the present study was to determine and characterize the lipid constituents/profile of the T. serratus venom utilizing liquid chromatography coupled with high-resolution mass spectrometry. Lipid species (164 in total) belonging to 3 different lipid categories, glycerophospholipids, sphingolipids, and glycerolipids, were identified. A further search on MetaCore/MetaDrug platform, which is based upon a manually curated database of molecular interactions, molecular pathways, gene-disease associations, chemical metabolism, and toxicity information, exhibited several metabolic pathways for 24 of previously identified lipid species, including activation of nuclear factor kappa B and oxidative stress pathways. Further several bioactive compounds, such as plasmalogens, lyso-platelet-activating factors, and sphingomyelins, associated with systemic responses triggered by T. serrulatus envenomation were detected. Finally, lipidomic data presented provide advanced and valuable information to better comprehend the mechanisms underlying the complex pathophysiology induced by T. serrulatus envenomation.
Collapse
Affiliation(s)
| | | | | | - Fernando Barbosa
- Departamento de Análise Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Lúcia Helena Faccioli
- Departamento de Análise Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
23
|
Silva de França F, Tambourgi DV. Hyaluronan breakdown by snake venom hyaluronidases: From toxins delivery to immunopathology. Front Immunol 2023; 14:1125899. [PMID: 37006255 PMCID: PMC10064005 DOI: 10.3389/fimmu.2023.1125899] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/14/2023] [Indexed: 03/19/2023] Open
Abstract
Snake venom enzymes have a broad range of molecular targets in plasma, tissues, and cells, among which hyaluronan (HA) is outstanding. HA is encountered in the extracellular matrix of diverse tissues and in the bloodstream, and its different chemical configurations dictate the diverse morphophysiological processes in which it participates. Hyaluronidases are highlighted among the enzymes involved in HA metabolism. This enzyme has been detected along the phylogenetic tree, suggesting that hyaluronidases exert multiple biological effects on different organisms. Hyaluronidases have been described in tissues, blood and snake venoms. Snake venom hyaluronidases (SVHYA) contribute to tissue destruction in envenomations and are called spreading factors since their action potentiates venom toxin delivery. Interestingly, SVHYA are clustered in Enzyme Class 3.2.1.35 together with mammalian hyaluronidases (HYAL). Both HYAL and SVHYA of Class 3.2.1.35 act upon HA, generating low molecular weight HA fragments (LMW-HA). LMW-HA generated by HYAL becomes a damage-associated molecular pattern that is recognized by Toll-like receptors 2 and 4, triggering cell signaling cascades culminating in innate and adaptive immune responses that are characterized by lipid mediator generation, interleukin production, chemokine upregulation, dendritic cell activation and T cell proliferation. In this review, aspects of the structures and functions of HA and hyaluronidases in both snake venoms and mammals are presented, and their activities are compared. In addition, the potential immunopathological consequences of HA degradation products generated after snakebite envenoming and their use as adjuvant to enhance venom toxin immunogenicity for antivenom production as well as envenomation prognostic biomarker are also discussed.
Collapse
|
24
|
Guerra-Duarte C, Saavedra-Langer R, Matavel A, Oliveira-Mendes BBR, Chavez-Olortegui C, Paiva ALB. Scorpion envenomation in Brazil: Current scenario and perspectives for containing an increasing health problem. PLoS Negl Trop Dis 2023; 17:e0011069. [PMID: 36757916 PMCID: PMC9910716 DOI: 10.1371/journal.pntd.0011069] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Opportunistic scorpion species can colonize urban environments, establishing high-density communities that enhance the chances of human accidents. This scenario has been taking place in Brazil, in which some Tityus species have taken city centers, causing an explosion in the number of scorpion envenoming cases. The characteristics of this scorpionism epidemic in Brazil is discussed in the present work. The number of Brazilian scorpion stings has surpassed 120,000 cases in 2017, and has been maintained above this number ever since, representing a more than 3-fold increase in 10 years, which was higher than the number of cases for most of the neglected tropical diseases in the country. The escalation in scorpionism cases is even higher in some regions of Brazil. Fortunately, the proportion of mild cases has also increased in the analyzed period, as well as the number of victims seeking for medical attention within the first hour after the accident. The species Tityus serrulatus, Tityus stigmurus, Tityus bahiensis, and Tityus obscurus are traditionally accountable for most of the scorpion accidents in different regions of Brazil, but other species deserve to be closely watched. Despite scorpionism being a notable health problem in Brazil, accident prevention and pest control regarding this venomous animal have not been properly addressed by the scientific community nor by policy makers. Therefore, this review also aims to point possible fields of research that could help to contain the aggravation of the current scorpionism landscape in Brazil.
Collapse
Affiliation(s)
- Clara Guerra-Duarte
- Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Rafael Saavedra-Langer
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alessandra Matavel
- Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | | | - Carlos Chavez-Olortegui
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
25
|
Cerni F, Oliveira I, Cordeiro F, Bordon K, Ferreira I, Monteiro W, Arantes E, Cunha T, Pucca M. The nociceptive response induced by different classes of Tityus serrulatus neurotoxins: The important role of Ts5 in venom-induced nociception. PLoS Negl Trop Dis 2023; 17:e0011057. [PMID: 36716327 PMCID: PMC9886245 DOI: 10.1371/journal.pntd.0011057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/23/2022] [Indexed: 02/01/2023] Open
Abstract
Scorpion sting envenomations (SSE) are feared by the intense pain that they produce in victims. Pain from SSE is triggered mainly by the presence of neurotoxins in the scorpion venom that modulates voltage-gated ion channels. In Brazil, SSE is mostly caused by Tityus serrulatus, popularly known as yellow scorpion. Here, we evaluated experimental spontaneous nociception induced by T. serrulatus venom as well as its isolated neurotoxins Ts1, Ts5, Ts6, Ts8, and Ts19 frag II, evidencing different degrees of pain behavior in mice. In addition, we developed a mice-derived polyclonal antibody targeting Ts5 able to neutralize the effect of this neurotoxin, showing that Ts5 presents epitopes capable of activating the immune response, which decreased considerably the nociception produced by the whole venom. This is the pioneer study to explore nociception using different classes of T. serrulatus neurotoxins on nociception (α-NaTx, β-NaTx, α-KTx, and β-KTx), targeting potassium and sodium voltage-gated channels, besides demonstrating that Ts5 plays an important role in the scorpion sting induced-pain.
Collapse
Affiliation(s)
- Felipe Cerni
- Health and Sciences Postgraduate Program, Federal University of Roraima, Boa Vista, Roraima, Brazil
| | - Isadora Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Francielle Cordeiro
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Karla Bordon
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Isabela Ferreira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Wuelton Monteiro
- Department of Teaching and Research, Dr. Heitor Vieira Dourado Tropical Medicine Foundation, Manaus, Amazonas, Brazil,Department of Medicine and Nursing, School of Health Sciences, Amazonas State University, Manaus, Amazonas, Brazil
| | - Eliane Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Manuela Pucca
- Health and Sciences Postgraduate Program, Federal University of Roraima, Boa Vista, Roraima, Brazil,Medical School, Federal University of Roraima, Boa Vista, Roraima, Brazil,* E-mail:
| |
Collapse
|
26
|
Design, synthesis, and biological evaluation of dual-target COX-2/5-LOX inhibitors for the treatment of inflammation. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02995-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
27
|
GPSM1 impairs metabolic homeostasis by controlling a pro-inflammatory pathway in macrophages. Nat Commun 2022; 13:7260. [PMID: 36434066 PMCID: PMC9700814 DOI: 10.1038/s41467-022-34998-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
G-protein-signaling modulator 1 (GPSM1) exhibits strong genetic association with Type 2 diabetes (T2D) and Body Mass Index in population studies. However, how GPSM1 carries out such control and in which types of cells are poorly understood. Here, we demonstrate that myeloid GPSM1 promotes metabolic inflammation to accelerate T2D and obesity development. Mice with myeloid-specific GPSM1 ablation are protected against high fat diet-induced insulin resistance, glucose dysregulation, and liver steatosis via repression of adipose tissue pro-inflammatory states. Mechanistically, GPSM1 deficiency mainly promotes TNFAIP3 transcription via the Gαi3/cAMP/PKA/CREB axis, thus inhibiting TLR4-induced NF-κB signaling in macrophages. In addition, we identify a small-molecule compound, AN-465/42243987, which suppresses the pro-inflammatory phenotype by inhibiting GPSM1 function, which could make it a candidate for metabolic therapy. Furthermore, GPSM1 expression is upregulated in visceral fat of individuals with obesity and is correlated with clinical metabolic traits. Overall, our findings identify macrophage GPSM1 as a link between metabolic inflammation and systemic homeostasis.
Collapse
|
28
|
Luo Q, Liu M, Tan Y, Chen J, Zhang W, Zhong S, Pan J, Zheng Q, Gong L, Su L, Jia Z, Dou X. Blockade of prostaglandin E2 receptor 4 ameliorates peritoneal dialysis-associated peritoneal fibrosis. Front Pharmacol 2022; 13:1004619. [DOI: 10.3389/fphar.2022.1004619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/28/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory responses in the peritoneum contribute to peritoneal dialysis (PD)-associated peritoneal fibrosis. Results of our previous study showed that increased microsomal prostaglandin E synthase-1-mediated production of prostaglandin E2 (PGE2) contributed to peritoneal fibrosis. However, the role of its downstream receptor in the progression of peritoneal fibrosis has not been established. Here, we examined the role of PGE2 receptor 4 (EP4) in the development of peritoneal fibrosis. EP4 was significantly upregulated in peritoneal tissues of PD patients with ultrafiltration failure, along with the presence of an enhanced inflammatory response. In vitro experiments showed that exposure to high glucose concentrations enhanced EP4 expression in rat peritoneal mesothelial cells (RPMCs). High-glucose–induced expression of inflammatory cytokines (monocyte chemoattractant protein-1, tumour necrosis factor α, and interleukin 1β) was significantly reduced in RPMCs treated with ONO-AE3-208, an EP4 receptor antagonist. ONO-AE3-208 also significantly decreased the expression of extracellular matrix proteins induced by high glucose concentrations. Furthermore, ONO-AE3-208 blunted activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome and phosphorylation of nuclear factor kappa B (NF-κB) (p-p65). To further investigate the functional role of EP4, ONO-AE3-208 was administrated for 4 weeks in a rat model of PD, the results of which showed that ONO-AE3-208 inhibited peritoneal fibrosis and improved peritoneal dysfunction. Additionally, inflammatory cytokines in the peritoneum of PD rats treated with ONO-AE3-208 were downregulated, in line with inhibition of the NLRP3 inflammasome and NF-κB phosphorylation. In conclusion, an EP4 antagonist reduced the development of peritoneal fibrosis, possibly by suppressing NLRP3 inflammasome- and p-p65–mediated inflammatory responses. Our findings suggest that an EP4 antagonist may be therapeutically beneficial for PD-associated peritoneal fibrosis.
Collapse
|
29
|
Paloschi MV, Boeno CN, Lopes JA, Rego CMA, Silva MDS, Santana HM, Serrath SN, Ikenohuchi YJ, Farias BJC, Felipin KP, Nery NM, Dos Reis VP, de Lima Lemos CT, Evangelista JR, da Silva Setúbal S, Soares AM, Zuliani JP. Reactive oxygen species-dependent-NLRP3 inflammasome activation in human neutrophils induced by l-amino acid oxidase derived from Calloselasma rhodostoma venom. Life Sci 2022; 308:120962. [PMID: 36113732 DOI: 10.1016/j.lfs.2022.120962] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/31/2022] [Accepted: 09/10/2022] [Indexed: 11/20/2022]
Abstract
l-Amino acid oxidase isolated from Calloselasma rhodostoma (Cr-LAAO) snake venom is a potent stimulus for neutrophil activation and production of inflammatory mediators, contributing to local inflammatory effects in victims of envenoming. Cr-LAAO triggered the activation of nicotinamide adenine dinucleotide phosphatase (NADPH) oxidase complex and protein kinase C (PKC)-α signaling protein for reactive oxygen species (ROS) production. This study aims to evaluate the ROS participation in the NLRP3 inflammasome complex activation in human neutrophil. Human neutrophils were isolated and stimulated for 1 or 2 h with RPMI (negative control), LPS (1 μg/mL, positive control) or Cr-LAAO (50 μg/mL). The neutrophil transcriptome was examined using the microarray technique, and RT-qPCR for confirmation of gene expression. Immunofluorescence assays for NLRP3, caspase-1, IL-1β and GSDMD proteins was performed by Western blot in the presence and/or absence of Apocynin, an inhibitor of NADPH oxidase. IL-1β release was also detected in the presence and/or absence of NLRP3, caspase-1 and NADPH oxidase inhibitors. Results showed that Cr-LAAO upregulated the expression of genes that participate in the NADPH oxidase complex formation and inflammasome assembly. NLRP3 was activated and accumulated in the cytosol forming punctas, indicating its activation. Gasdermin D was not cleaved but lactate dehydrogenase was released. Furthermore, ROS inhibition decreased the expression of NLRP3 inflammasome complex proteins, as observed by protein expression in the presence and/or absence of apocynin, an NADPH oxidase inhibitor. IL-1β was also released, and pharmacological inhibition of NLRP3, caspase-1, and ROS reduced the amount of released cytokine. This is the first report demonstrating the activation of the NLRP3 inflammasome complex via ROS generation by Cr-LAAO, which may lead to the development of local inflammatory effects observed in snakebite victims.
Collapse
Affiliation(s)
- Mauro Valentino Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Charles Nunes Boeno
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Jéssica Amaral Lopes
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Cristina Matiele Alves Rego
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Milena Daniela Souza Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Hallison Mota Santana
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Suzanne Nery Serrath
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Yoda Janaina Ikenohuchi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Braz Junior Campos Farias
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Kátia Paula Felipin
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Neriane Monteiro Nery
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Valdison Pereira Dos Reis
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Caleb Torres de Lima Lemos
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Jaina Rodrigues Evangelista
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Sulamita da Silva Setúbal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Andreimar Martins Soares
- Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia e Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Juliana Pavan Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia e Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
30
|
Boeno CN, Paloschi MV, Lopes JA, Souza Silva MD, Evangelista JR, Dos Reis VP, da S Setúbal S, Soares AM, Zuliani JP. Dynamics of action of a Lys-49 and an Asp-49 PLA 2s on inflammasome NLRP3 activation in murine macrophages. Int Immunopharmacol 2022; 112:109194. [PMID: 36041255 DOI: 10.1016/j.intimp.2022.109194] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/08/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
Phospholipases A2 (PLA2s) are proteins found in snake venoms with hemolytic, anticoagulant, myotoxic, edematogenic, bactericidal and inflammatory actions. In Bothrops jararacussu snake venom were isolated a Lys49-PLA2 (BthTX-I) and an Asp49-PLA2 (BthTX-II) with myotoxic and inflammatory actions. Both PLA2s can activate the NLRP3 inflammasome, an intracytoplasmic platform that recognizes molecules released when tissue is damaged liberating IL-1β that contributes to the inflammatory response observed in envenoming. The dynamic of action of BthTX-I and BthTX-II in both thioglycollate (TG)-elicited macrophages and C2C12 myoblasts and the involvement of EP1 and EP2 receptors, and PGE2 in NLRP3 inflammasome activation were evaluated. Both toxins induced PGE2 liberation and inflammasome components (NLRP3, Caspase-1, ASC, IL-1β, and IL18), IL-6, P2X7, COX-1, COX-2, EP2 and EP4 gene expression in TG-elicited macrophages but not in C2C12 myoblasts. EP2 (PF04418948) and EP4 (GW627368X) inhibitors abolished this effect. Both PLA2s also induced NLRP3 inflammasome protein expression that was abolished with the inhibitors used. Immunofluorescence and IL-1β assays confirmed the NLRP3 activation in TG-elicited macrophages with the participation of both EP2 and EP4 receptors confirming their involvement in this effect. All in all, BthTX-I and BthTX-II activate macrophages and induce the NLRP3 inflammasome complex activation with the participation of the PGE2 via COX pathway and EP2 and EP4, both PGE2 receptors, contributing to the local inflammatory effects observed in envenoming.
Collapse
Affiliation(s)
- Charles N Boeno
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Mauro V Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Jéssica A Lopes
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Milena D Souza Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Jaína R Evangelista
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Valdison P Dos Reis
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Sulamita da S Setúbal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Andreimar M Soares
- Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia e Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Juliana P Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia e Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
31
|
Pérez MM, Pimentel VE, Fuzo CA, da Silva-Neto PV, Toro DM, Fraga-Silva TFC, Gardinassi LG, Oliveira CNS, Souza COS, Torre-Neto NT, de Carvalho JCS, De Leo TC, Nardini V, Feitosa MR, Parra RS, da Rocha JJR, Feres O, Vilar FC, Gaspar GG, Constant LF, Ostini FM, Degiovani AM, Amorim AP, Viana AL, Fernandes APM, Maruyama SR, Russo EMS, Santos IKFM, Bonato VLD, Cardoso CRB, Sorgi CA, Dias-Baruffi M, Faccioli LH. Acetylcholine, Fatty Acids, and Lipid Mediators Are Linked to COVID-19 Severity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:250-261. [PMID: 35768148 DOI: 10.4049/jimmunol.2200079] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
Abstract
Lipid and cholinergic mediators are inflammatory regulators, but their role in the immunopathology of COVID-19 is still unclear. Here, we used human blood and tracheal aspirate (TA) to investigate whether acetylcholine (Ach), fatty acids (FAs), and their derived lipid mediators (LMs) are associated with COVID-19 severity. First, we analyzed the perturbation profile induced by SARS-CoV-2 infection in the transcriptional profile of genes related to the ACh and FA/LM pathways. Blood and TA were used for metabolomic and lipidomic analyses and for quantification of leukocytes, cytokines, and ACh. Differential expression and coexpression gene network data revealed a unique transcriptional profile associated with ACh and FA/LM production, release, and cellular signaling. Transcriptomic data were corroborated by laboratory findings: SARS-CoV-2 infection increased plasma and TA levels of arachidonic acid, 5-hydroxy-6E,8Z,11Z,14Z-eicosatetraenoic acid, 11-hydroxy-5Z,8Z,12E,14Z-eicosatetraenoic acid, and ACh. TA samples also exhibited high levels of PGE2, thromboxane B2, 12-oxo-5Z,8Z,10E,14Z-eicosatetraenoic acid, and 6-trans-leukotriene B4 Bioinformatics and experimental approaches demonstrated robust correlation between transcriptional profile in Ach and FA/LM pathways and parameters of severe COVID-19. As expected, the increased neutrophil-to-lymphocyte ratio, neutrophil counts, and cytokine levels (IL-6, IL-10, IL-1β, and IL-8) correlated with worse clinical scores. Glucocorticoids protected severe and critical patients and correlated with reduced Ach levels in plasma and TA samples. We demonstrated that pulmonary and systemic hyperinflammation in severe COVID-19 are associated with high levels of Ach and FA/LM. Glucocorticoids favored the survival of patients with severe/critical disease, and this effect was associated with a reduction in ACh levels.
Collapse
Affiliation(s)
- Malena M Pérez
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Vinícius E Pimentel
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto São Paulo, Brazil
| | - Carlos A Fuzo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Pedro V da Silva-Neto
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Diana M Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Thais F C Fraga-Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz G Gardinassi
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Camilla N S Oliveira
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto São Paulo, Brazil
| | - Camila O S Souza
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto São Paulo, Brazil
| | - Nicola T Torre-Neto
- Departamento de Química. Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jonatan C S de Carvalho
- Departamento de Química. Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thais C De Leo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Viviani Nardini
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marley R Feitosa
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rogerio S Parra
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José J R da Rocha
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Omar Feres
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando C Vilar
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Gilberto G Gaspar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Leticia F Constant
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Fátima M Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Augusto M Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Alessandro P Amorim
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Angelina L Viana
- Departamento de Enfermagem Materno-Infantil e Saúde Pública, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana P M Fernandes
- Departamento de Enfermagem Geral e Especializada, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sandra R Maruyama
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Elisa M S Russo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Isabel K F M Santos
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Vânia L D Bonato
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Cristina R B Cardoso
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos A Sorgi
- Departamento de Química. Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil;
| | | |
Collapse
|
32
|
Beltrán-García J, Osca-Verdegal R, Pérez-Cremades D, Novella S, Hermenegildo C, Pallardó FV, García-Giménez JL. Extracellular Histones Activate Endothelial NLRP3 Inflammasome and are Associated with a Severe Sepsis Phenotype. J Inflamm Res 2022; 15:4217-4238. [PMID: 35915852 PMCID: PMC9338392 DOI: 10.2147/jir.s363693] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/29/2022] [Indexed: 12/27/2022] Open
Affiliation(s)
- Jesús Beltrán-García
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Rebeca Osca-Verdegal
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Daniel Pérez-Cremades
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Susana Novella
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Carlos Hermenegildo
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - Federico V Pallardó
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
| | - José Luis García-Giménez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, Spain
- Correspondence: José Luis García-Giménez, Departamento de Fisiología, Facultad de Medicina y Odontología, Universitat de València, València, 46010, Spain, Tel +34 963 864 646, Email
| |
Collapse
|
33
|
Galvão-Lima LJ, Zambuzi FA, Soares LS, Fontanari C, Meireles AFG, Brauer VS, Faccioli LH, Gama L, Figueiredo LTM, Bou-Habib DC, Frantz FG. HIV-1 Gag and Vpr impair the inflammasome activation and contribute to the establishment of chronic infection in human primary macrophages. Mol Immunol 2022; 148:68-80. [PMID: 35659727 DOI: 10.1016/j.molimm.2022.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023]
Abstract
The successful establishment of HIV-1 infection is related to inflammasome blocking or inactivation, which can result in the viral evasion of the immune responses and formation of reservoirs in several tissues. In this sense, we aimed to evaluate the viral and cellular mechanisms activated during HIV-1 infection in human primary macrophages that allow an effective viral replication in these cells. We found that resting HIV-1-infected macrophages, but not those activated in classical or alternative patterns, released IL-1β and other pro-inflammatory cytokines, and showed increased CXCL10 expression, without changes in the NLRP3, AIM2 or RIG-I inflammasome pathways. Also, similar levels of Casp-1, phosphorylated NF-κB (p65) and NLRP3 proteins were found in uninfected and HIV-1-infected macrophages. Likewise, no alterations were detected in ASC specks released in the culture supernatant after HIV-1 infection, suggesting that macrophages remain viable after infection. Using in silico prediction studies, we found that the HIV-1 proteins Gag and Vpr interact with several host proteins. Comparable levels of trans-LTB4 were found in the supernatants of uninfected and HIV-1-infected macrophages, whereas ROS production was impaired in infected cells, which was not reversed after the PMA stimulus. Immunofluorescence analysis showed structural alterations in the mitochondrial architecture and an increase of BIM in the cytoplasm of infected cells. Our data suggest that HIV-1 proteins Gag and Vpr, through interacting with cellular proteins in the early steps of infection, preclude the inflammasome activation and the development of effective immune responses, thus allowing the establishment of the infection.
Collapse
Affiliation(s)
- Leonardo J Galvão-Lima
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil; Laboratory of Technological Innovation in Health (LAIS), Hospital Universitário Onofre Lopes, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | - Fabiana A Zambuzi
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Luana S Soares
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Caroline Fontanari
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Aline F Galvão Meireles
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Verônica S Brauer
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Lúcia H Faccioli
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Lúcio Gama
- Retrovirus Lab, Johns Hopkins University - School of Medicine, Baltimore, MD, USA; Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Luiz T M Figueiredo
- Virology Research Center, Medical School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| | - Fabiani G Frantz
- School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
34
|
Ghezellou P, Jakob K, Atashi J, Ghassempour A, Spengler B. Mass-Spectrometry-Based Lipidome and Proteome Profiling of Hottentotta saulcyi (Scorpiones: Buthidae) Venom. Toxins (Basel) 2022; 14:toxins14060370. [PMID: 35737031 PMCID: PMC9228814 DOI: 10.3390/toxins14060370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/10/2022] Open
Abstract
Scorpion venom is a complex secretory mixture of components with potential biological and physiological properties that attracted many researchers due to promising applications from clinical and pharmacological perspectives. In this study, we investigated the venom of the Iranian scorpion Hottentotta saulcyi (Simon, 1880) by applying mass-spectrometry-based proteomic and lipidomic approaches to assess the diversity of components present in the venom. The data revealed that the venom’s proteome composition is largely dominated by Na+- and K+-channel-impairing toxic peptides, following the enzymatic and non-enzymatic protein families, e.g., angiotensin-converting enzyme, serine protease, metalloprotease, hyaluronidase, carboxypeptidase, and cysteine-rich secretory peptide. Furthermore, lipids comprise ~1.2% of the dry weight of the crude venom. Phospholipids, ether-phospholipids, oxidized-phospholipids, triacylglycerol, cardiolipins, very-long-chain sphingomyelins, and ceramides were the most intensely detected lipid species in the scorpion venom, may acting either independently or synergistically during the envenomation alongside proteins and peptides. The results provide detailed information on the chemical makeup of the venom, helping to improve our understanding of biological molecules present in it, leading to a better insight of the medical significance of the venom, and improving the medical care of patients suffering from scorpion accidents in the relevant regions such as Iran, Iraq, Turkey, and Afghanistan.
Collapse
Affiliation(s)
- Parviz Ghezellou
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany;
- Correspondence: (P.G.); (B.S.)
| | - Kevin Jakob
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Javad Atashi
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983969411, Iran; (J.A.); (A.G.)
| | - Alireza Ghassempour
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983969411, Iran; (J.A.); (A.G.)
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany;
- Correspondence: (P.G.); (B.S.)
| |
Collapse
|
35
|
Fang M, Tang X, Zhang J, Liao Z, Wang G, Cheng R, Zhang Z, Zhao H, Wang J, Tan Z, Kamau PM, Lu Q, Liu Q, Deng G, Lai R. An inhibitor of leukotriene-A 4 hydrolase from bat salivary glands facilitates virus infection. Proc Natl Acad Sci U S A 2022; 119:e2110647119. [PMID: 35238649 PMCID: PMC8915838 DOI: 10.1073/pnas.2110647119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/20/2021] [Indexed: 12/23/2022] Open
Abstract
SignificanceAn immunosuppressant protein (MTX), which facilitates virus infection by inhibiting leukotriene A4 hydrolase (LTA4H) to produce the lipid chemoattractant leukotriene B4 (LTB4), was identified and characterized from the submandibular salivary glands of the bat Myotis pilosus. To the best of our knowledge, this is a report of an endogenous LTA4H inhibitor in animals. MTX was highly concentrated in the bat salivary glands, suggesting a mechanism for the generation of immunological privilege and immune tolerance and providing evidence of viral shedding through oral secretions. Moreover, given that the immunosuppressant MTX selectively inhibited the proinflammatory activity of LTA4H, without affecting its antiinflammatory activity, MTX might be a potential candidate for the development of antiinflammatory drugs by targeting the LTA4-LTA4H-LTB4 inflammatory axis.
Collapse
Affiliation(s)
- Mingqian Fang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
| | - Xiaopeng Tang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
| | - Juan Zhang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhiyi Liao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gan Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Ruomei Cheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiye Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Hongwen Zhao
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jing Wang
- Department of Laboratory Diagnosis, Chongqing Public Health Medical Center, Public Health Hospital of Southwest University, Shapingba District, Chongqing 400038, China
| | - Zhaoxia Tan
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peter Muiruri Kamau
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Qiumin Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Qi Liu
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guohong Deng
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| |
Collapse
|
36
|
de Macedo LH, Souza COS, Gardinassi LG, Faccioli LH. CD14 regulates the metabolomic profiles of distinct macrophage subsets under steady and activated states. Immunobiology 2022; 227:152191. [DOI: 10.1016/j.imbio.2022.152191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 11/05/2022]
|
37
|
He X, Wang J, Wang Q, Liu J, Yang X, He L, Hu H, Zeng S, Yu L, Qiu Y, Lou Y. P38 MAPK, NF-κB, and JAK-STAT3 Signaling Pathways Involved in Capecitabine-Induced Hand-Foot Syndrome via Interleukin 6 or Interleukin 8 Abnormal Expression. Chem Res Toxicol 2022; 35:422-430. [PMID: 35147423 DOI: 10.1021/acs.chemrestox.1c00317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hand-foot syndrome (HFS) is a major adverse reaction to capecitabine (CAP). The exact pathogenesis of this disease remains unclear. In this study, metabolomics combined with cell RNA sequencing was used to study the mechanisms of CAP-induced HFS. The murine model of HFS was constructed by intragastric administration of CAP or its metabolites. Quantitative reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assays were used to verify the mechanisms. Metabolomics showed the phosphatidylinositol signaling pathway and amino acid and fatty acid metabolism to be the major metabolic alterations related to the occurrence of HFS. Transcriptomics profiles further revealed that the cytokine-cytokine receptor interaction, IL17 signaling pathway, Toll-like receptor signaling pathway, arachidonic acid metabolism, MAPK signaling pathway, and JAK-STAT3 signaling pathway were the vital steps in skin toxicity induced by CAP or its metabolites. We also verified that the inflammation mechanisms were primarily mediated by the abnormal expression of interleukin (IL) 6 or IL8 and not exclusively by COX-2 overexpression. Finally, the P38 MAPK, NF-κB, and JAK-STAT3 signaling pathways, which mediate high levels of expression of IL6 or IL8, were identified as potential pathways underlying CAP-induced HFS.
Collapse
Affiliation(s)
- Xiaoying He
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Jiali Wang
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Qian Wang
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Jing Liu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Xi Yang
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Lingjuan He
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Haihong Hu
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Su Zeng
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Lushan Yu
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| | - Yan Lou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People's Republic of China
| |
Collapse
|
38
|
Elrayess RA, Mohallal ME, Mobarak YM, Ebaid HM, Haywood-Small S, Miller K, Strong PN, Abdel-Rahman MA. Scorpion Venom Antimicrobial Peptides Induce Caspase-1 Dependant Pyroptotic Cell Death. Front Pharmacol 2022; 12:788874. [PMID: 35082671 PMCID: PMC8784870 DOI: 10.3389/fphar.2021.788874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022] Open
Abstract
Within the last decade, several peptides have been identified according to their ability to inhibit the growth of microbial pathogens. These antimicrobial peptides (AMPs) are a part of the innate immune system of all living organisms. Many studies on their effects on prokaryotic microorganisms have been reported; some of these peptides have cytotoxic properties although the molecular mechanisms underlying their activity on eukaryotic cells remain poorly understood. Smp24 and Smp43 are novel cationic AMPs which were identified from the venom of the Egyptian scorpion Scorpio maurus palmatus. Smp24 and Smp43 showed potent activity against both Gram-positive and Gram-negative bacteria as well as fungi. Here we describe cytotoxicity of these peptides towards two acute leukaemia cell lines (myeloid (KG1-a) and lymphoid (CCRF-CEM) leukaemia cell lines) and three non-tumour cell lines CD34+ (hematopoietic stem progenitor from cord blood), HRECs (human renal epithelial cells) and HaCaT (human skin keratinocytes). Smp24 and Smp43 (4–256 µg/ml) decreased the viability of all cell lines, although HaCaT cells were markedly less sensitive. With the exception HaCaT cells, the caspase-1 gene was uniquely up-regulated in all cell lines studied. However, all cell lines showed an increase in downstream interleukin-1β (IL-1β) expression. Transmission electron microscope studies revealed the formation of cell membrane blebs and the appearance of autolysosomes and lipid droplets in all cell lines; KG1-a leukemia cells also showed the unique appearance of glycogen deposits. Our results reveal a novel mechanism of action for scorpion venom AMPs, activating a cascade of events leading to cell death through a programmed pyroptotic mechanism.
Collapse
Affiliation(s)
- Ranwa A Elrayess
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom.,Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Mahmoud E Mohallal
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Yomn M Mobarak
- Zoology Department, Faculty of Science, Suez University, Suez, Egypt
| | - Hala M Ebaid
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Sarah Haywood-Small
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Keith Miller
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Peter N Strong
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | | |
Collapse
|
39
|
Local inflammatory mediators alterations induced by Daboia siamensis venom. Toxicon X 2021; 12:100085. [PMID: 34693275 PMCID: PMC8517603 DOI: 10.1016/j.toxcx.2021.100085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 07/15/2021] [Accepted: 09/28/2021] [Indexed: 11/27/2022] Open
Abstract
The ability of Russell's viper (Daboia siamensis) venom (total RVV) and phospholipase A2 (purified PLA2) to induce the local pathological effects were investigated by the local inflammatory events and the release of inflammatory mediators. Both 0.5 μg of total RVV/mouse and 0.15 μg of purified PLA2/mouse were administered via intra-peritoneal injection. After 30 min, 1 h, 2 h, and 4 h incubation time, the peritoneal cavity was flooded with normal saline and the total leukocytes were collected. The eicosanoids (lipid mediators) and the leukocyte expression of cyclooxygenase (COX-1 and COX-2) were investigated by ELISA assay and western blotting, respectively. The amounts of total leukocytes were increased from 30 min to 2 h, then decreased at 4 h, by both total RVV and purified PLA2. Both treatments also induced the expression of COX-2 which was increased at 2 h and then decreased at 4 h, whereas only purified PLA2 induced the expression level of a COX-1 protein which was increased at 30 min, then constantly expressed until 4 h. In addition, total RVV and purified PLA2 caused the release of the eicosanoids; PGE2, TXB2, and LTB4, which reached the peak after 2 h. The findings of this study indicate that purified PLA2 has the potential effects to induce the local inflammation relating the amounts of leukocytes cells, lipid mediators and COX-2 more than the total RVV. Purified phospholipase A2 or the venom could induce eicosanoids and cyclooxygenase-2 expression relating to leukocytes cells. Thromboxane B2 could be the important mediator induced by Russell's viper venom and purified phospholipase A2. Russell's viper venom and purified phospholipase A2 involved the cyclooxygenase-2 expression, but not cyclooxygenase-1. The purified phospholipase A2 showed more predominant inflammatory response at site than total Russell's viper venom.
Collapse
|
40
|
Ferreira JC, Reis MB, Coelho GDP, Gastaldello GH, Peti APF, Rodrigues DM, Bastos JK, Campo VL, Sorgi CA, Faccioli LH, Gardinassi LG, Tefé-Silva C, Zoccal KF. Baccharin and p-coumaric acid from green propolis mitigate inflammation by modulating the production of cytokines and eicosanoids. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114255. [PMID: 34062248 DOI: 10.1016/j.jep.2021.114255] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Green propolis is produced by Apis mellifera honeybees using Baccharis dracunculifolia D.C. (Asteraceae) as substrate. This Southern Brazilian native plant and green propolis have been used in traditional medicine to treat gastric diseases, inflammation and liver disorders. AIM OF THE STUDY Investigate the effects of baccharin (Bac) or p-coumaric acid (pCA) isolated from B. dracunculifolia D.C. (Asteraceae) over the inflammation induced by lipopolysaccharide (LPS) in vivo. MATERIALS AND METHODS Inflammation was induced by LPS injection into air-pouches in mice, which were subsequently treated with Bac or pCA. Lavage fluid was collected from air pouches for the quantification of cellular influx via microscopy, and quantification of inflammatory mediators via colorimetric methods, ELISA and liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS LPS-induced inflammation increased cellular influx and increased the levels of parameters related to vascular permeability and edema formation, such as nitric oxide (NO) and protein extravasation. Moreover, LPS increased the levels of cytokines and eicosanoids in the air-pouches. Importantly, both Bac and pCA suppressed the infiltration of neutrophils, production of NO and protein extravasation. Notably, the compounds promote differential regulation of cytokine and eicosanoid production. CONCLUSIONS Our results suggest that Bac from green propolis directly affects inflammation by inhibiting the production of cytokines and eicosanoids, while pCA may exert direct, but also indirect effects on inflammation by stimulating the production of regulatory effectors such as interkeukin-10 in vivo.
Collapse
Affiliation(s)
- Juliana C Ferreira
- Centro Universitário Barão de Mauá (CBM), Ribeirão Preto, São Paulo, Brazil.
| | - Mouzarllem B Reis
- Centro Universitário Barão de Mauá (CBM), Ribeirão Preto, São Paulo, Brazil.
| | - Giovanna D P Coelho
- Centro Universitário Barão de Mauá (CBM), Ribeirão Preto, São Paulo, Brazil.
| | | | - Ana Paula F Peti
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Débora M Rodrigues
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Jairo K Bastos
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Vanessa L Campo
- Centro Universitário Barão de Mauá (CBM), Ribeirão Preto, São Paulo, Brazil; Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Carlos A Sorgi
- Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Lúcia H Faccioli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Luiz G Gardinassi
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil.
| | | | - Karina F Zoccal
- Centro Universitário Barão de Mauá (CBM), Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
41
|
Martins JG, Santos GC, Procópio REDL, Arantes EC, Bordon KDCF. Scorpion species of medical importance in the Brazilian Amazon: a review to identify knowledge gaps. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20210012. [PMID: 34589120 PMCID: PMC8452272 DOI: 10.1590/1678-9199-jvatitd-2021-0012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/09/2021] [Indexed: 12/24/2022] Open
Abstract
Scorpionism is a relevant medical condition in Brazil. It is responsible for most accidents involving venomous animals in the country, which leads to severe symptoms that can evolve to death. In recent years, an increase of almost 50% in the incidence of scorpionism has been observed in the Northern Region, where the highest severity of envenoming has been notified since the beginning of the 21st century. This review aims to provide an in-depth assessment of public data and reports on symptoms and epidemiology of envenoming, ecological aspects of scorpions, and characterization of venoms and toxins to access the gaps that need to be filled in the knowledge of the scorpion species of medical importance from the Brazilian Amazon. A systematic search using the string words "Amazon" and "scorpion" was performed on 11 databases. No restriction on date, language or status of the publication was applied. Reports not related to the Brazilian Amazon were excluded. Therefore, 88 studies remained. It is shown that populations of scorpions of medical importance, even of the same species, may present significant toxic variations peculiar to some regions in the Brazilian Amazon, and commercial scorpion antivenoms were not able to shorten the intensity and duration of neurological manifestations in patients stung by T. silvestris, T. apiacas or T. obscurus. It is also highlighted that the toxins responsible for triggering these alterations have not been elucidated yet and this is a fruitful field for the development of more efficient antivenoms. Furthermore, the geographic distribution of scorpions of the genus Tityus in the Brazilian Amazon was revised and updated. The cumulative and detailed information provided in this review may help physicians and scientists interested in scorpionism in the Brazilian Amazon.
Collapse
Affiliation(s)
- Jonas Gama Martins
- Graduate Program in Genetics, Conservation and Evolutionary Biology
(PPG GCBEv), National Institute for Amazon Research (INPA), Manaus, AM,
Brazil
| | - Gabrielle Cristina Santos
- Department of BioMolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Rudi Emerson de Lima Procópio
- Graduate Program in Biotechnology and Natural Resources of Amazon,
University of the State of Amazonas (UEA), Manaus, AM, Brazil
| | - Eliane Candiani Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Karla de Castro Figueiredo Bordon
- Department of BioMolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| |
Collapse
|
42
|
Souza COS, Ketelut-Carneiro N, Milanezi CM, Faccioli LH, Gardinassi LG, Silva JS. NLRC4 inhibits NLRP3 inflammasome and abrogates effective antifungal CD8 + T cell responses. iScience 2021; 24:102548. [PMID: 34142053 PMCID: PMC8184506 DOI: 10.1016/j.isci.2021.102548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/06/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022] Open
Abstract
The recognition of fungi by intracellular NOD-like receptors (NLRs) induces inflammasome assembly and activation. Although the NLRC4 inflammasome has been extensively studied in bacterial infections, its role during fungal infections is unclear. Paracoccidioidomycosis (PCM) is a pathogenic fungal disease caused by Paracoccidioides brasiliensis. Here, we show that NLRC4 confers susceptibility to experimental PCM by regulating NLRP3-dependent cytokine production and thus protective effector mechanisms. Early after infection, NLRC4 suppresses prostaglandin E2 production, and consequently reduces interleukin (IL)-1β release by macrophages and dendritic cells in the lungs. IL-1β is required to control fungal replication via induction of the nitric oxide synthase 2 (NOS2) pathway. At a later stage of the disease, NLRC4 impacts IL-18 release, dampening robust CD8+IFN-γ+ T cell responses and enhancing mortality of mice. These findings demonstrate that NLRC4 promotes disease by regulating the production of inflammatory cytokines and cellular responses that depend on the NLRP3 inflammasome activity. NLRC4 promotes susceptibility to a highly pathogenic fungus. NLRC4 regulates NLRP3 activity. NLRC4 inhibits early NLRP3/IL-1β/NOS2/NO axis and promotes fungal replication. NLRC4 dampens late IL-18 production, suppressing CD8+IFN-γ+ T cell responses.
Collapse
Affiliation(s)
- Camila O S Souza
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Natália Ketelut-Carneiro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Program in Innate Immunity, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Cristiane M Milanezi
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lúcia H Faccioli
- Department of Clinical Analyses, Toxicology and Bromatological Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luiz G Gardinassi
- Department of Biosciences and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Platform, Ribeirão Preto, SP, Brazil
| |
Collapse
|
43
|
Jimenez-Duran G, Triantafilou M. Metabolic regulators of enigmatic inflammasomes in autoimmune diseases and crosstalk with innate immune receptors. Immunology 2021; 163:348-362. [PMID: 33682108 PMCID: PMC8274167 DOI: 10.1111/imm.13326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
Nucleotide‐binding domain and leucine‐rich repeat receptor (NLR)‐mediated inflammasome activation is important in host response to microbes, danger‐associated molecular patterns (DAMPs) and metabolic disease. Some NLRs have been shown to interact with distinct cell metabolic pathways and cause negative regulation, tumorigenesis and autoimmune disorders, interacting with multiple innate immune receptors to modulate disease. NLR activation is therefore crucial in host response and in the regulation of metabolic pathways that can trigger a wide range of immunometabolic diseases or syndromes. However, the exact mode by which some of the less well‐studied NLR inflammasomes are activated, interact with other metabolites and immune receptors, and the role they play in the progression of metabolic diseases is still not fully elucidated. In this study, we review up‐to‐date evidence regarding NLR function in metabolic pathways and the interplay with other immune receptors involved in GPCR signalling, gut microbiota and the complement system, in order to gain a better understanding of its link to disease processes.
Collapse
Affiliation(s)
- Gisela Jimenez-Duran
- Immunology Network, GlaxoSmithKline, Stevenage, UK.,Institute of Infection and Immunity, School of Medicine, University Hospital of Wales, Cardiff University, Cardiff, UK
| | - Martha Triantafilou
- Immunology Network, GlaxoSmithKline, Stevenage, UK.,Institute of Infection and Immunity, School of Medicine, University Hospital of Wales, Cardiff University, Cardiff, UK
| |
Collapse
|
44
|
Silva de França F, Villas-Boas IM, Cogliati B, Woodruff TM, Reis EDS, Lambris JD, Tambourgi DV. C5a-C5aR1 Axis Activation Drives Envenomation Immunopathology by the Snake Naja annulifera. Front Immunol 2021; 12:652242. [PMID: 33936074 PMCID: PMC8082402 DOI: 10.3389/fimmu.2021.652242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 12/03/2022] Open
Abstract
Systemic complement activation drives a plethora of pathological conditions, but its role in snake envenoming remains obscure. Here, we explored complement's contribution to the physiopathogenesis of Naja annulifera envenomation. We found that N. annulifera venom promoted the generation of C3a, C4a, C5a, and the soluble Terminal Complement Complex (sTCC) mediated by the action of snake venom metalloproteinases. N. annulifera venom also induced the release of lipid mediators and chemokines in a human whole-blood model. This release was complement-mediated, since C3/C3b and C5a Receptor 1 (C5aR1) inhibition mitigated the effects. In an experimental BALB/c mouse model of envenomation, N. annulifera venom promoted lipid mediator and chemokine production, neutrophil influx, and swelling at the injection site in a C5a-C5aR1 axis-dependent manner. N. annulifera venom induced systemic complementopathy and increased interleukin and chemokine production, leukocytosis, and acute lung injury (ALI). Inhibition of C5aR1 with the cyclic peptide antagonist PMX205 rescued mice from these systemic reactions and abrogated ALI development. These data reveal hitherto unrecognized roles for complement in envenomation physiopathogenesis, making complement an interesting therapeutic target in envenomation by N. annulifera and possibly by other snake venoms.
Collapse
Affiliation(s)
| | | | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Trent M. Woodruff
- Neuroinflammation Laboratory, School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Edimara da Silva Reis
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
45
|
Xie S, Qi X, Wu Q, Wei L, Zhang M, Xing Y, Shi W, Chen S, Zeng X, Wang S, Guo H, Deng W. Inhibition of 5-lipoxygenase is associated with downregulation of the leukotriene B4 receptor 1/ Interleukin-12p35 pathway and ameliorates sepsis-induced myocardial injury. Free Radic Biol Med 2021; 166:348-357. [PMID: 33705958 DOI: 10.1016/j.freeradbiomed.2021.02.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 11/21/2022]
Abstract
Sepsis rapidly contributed to multiorgan failure affecting most commonly of the cardiovascular and respiratory systems and yet there were no effective therapies. The current study aimed at providing evidence on the cardioprotection of suppression of 5-Lipoxygenase (5-Lox) and identifying the possible mechanism in the mouse model of sepsis. The cecal ligation-perforation (CLP) model was applied to C57BL/6 wild-type (WT) and 5-Lox deficient (5-Lox-/-) mice to induce sepsis. 5-Lox expression was up-regulated in mouse myocardium and leukotriene B4 (LTB4) level was increased in serum after sepsis. Subsequently, we utilized a recombinant adenoviral expression vector (rAAV9) to overexpress Alox5 gene in adult mice. Compared to WT mice, 5-Lox overexpression accelerated CLP-induced myocardial injury and cardiac dysfunction. Oppositely, 5-Lox deficiency offered protection against myocardial injury in a mouse model of sepsis and attenuated sepsis-mediated inflammation, oxidative stress and apoptosis in the mouse heart. Mechanically, 5-Lox promoted LTB4 production, which in turn contributed to the activation of leukotriene B4 receptor 1 (BLT1)/interleukin-12p35 (IL-12p35) pathway and enhanced M1 macrophage polarization. However, the suppression of BLT1 by either gene mutation or antagonist U75302 significantly inhibited the adverse effect of 5-Lox in sepsis. Further study demonstrated that pharmacological inhibition of 5-Lox prevented CLP-induced septic cardiomyopathy (SCM). Our study identified 5-Lox exacerbated sepsis-associated myocardial injury through activation of LTB4 production and promoting BLT1/IL-12p35 pathway. Hence, inhibition of 5-Lox may be a potential candidate strategy for septic cardiac dysfunction treatment.
Collapse
Affiliation(s)
- Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Xiping Qi
- Department of Transfusion, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No.1 Hospital), Wuhan, 430060, China
| | - Qingqing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Li Wei
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Yun Xing
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Wenke Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Si Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Xiaofeng Zeng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Shasha Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Haipeng Guo
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, 250012, China; Department of Critical Care Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| |
Collapse
|
46
|
Ranéia E Silva PA, de Lima DS, Mesquita Luiz JP, Câmara NOS, Alves-Filho JCF, Pontillo A, Bortoluci KR, Faquim-Mauro EL. Inflammatory effect of Bothropstoxin-I from Bothrops jararacussu venom mediated by NLRP3 inflammasome involves ATP and P2X7 receptor. Clin Sci (Lond) 2021; 135:687-701. [PMID: 33620070 DOI: 10.1042/cs20201419] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Muscle tissue damage is one of the local effects described in bothropic envenomations. Bothropstoxin-I (BthTX-I), from Bothrops jararacussu venom, is a K49-phospholipase A2 (PLA2) that induces a massive muscle tissue injury, and, consequently, local inflammatory reaction. The NLRP3 inflammasome is a sensor that triggers inflammation by activating caspase 1 and releasing interleukin (IL)-1β and/or inducing pyroptotic cell death in response to tissue damage. We, therefore, aimed to address activation of NLRP3 inflammasome by BthTX-I-associated injury and the mechanism involved in this process. Intramuscular injection of BthTX-I results in infiltration of neutrophils and macrophages in gastrocnemius muscle, which is reduced in NLRP3- and Caspase-1-deficient mice. The in vitro IL-1β production induced by BthTX-I in peritoneal macrophages (PMs) requires caspase 1/11, ASC and NLRP3 and is dependent on adenosine 5'-triphosphate (ATP)-induced K+ efflux and P2X7 receptor (P2X7R). BthTX-I induces a dramatic release of ATP from C2C12 myotubes, therefore representing the major mechanism for P2X7R-dependent inflammasome activation in macrophages. A similar result was obtained when human monocyte-derived macrophages (HMDMs) were treated with BthTX-I. These findings demonstrated the inflammatory effect of BthTX-I on muscle tissue, pointing out a role for the ATP released by damaged cells for the NLRP3 activation on macrophages, contributing to the understanding of the microenvironment of the tissue damage of the Bothrops envenomation.
Collapse
Affiliation(s)
- Priscila Andrade Ranéia E Silva
- Laboratory of Immunopathology, Butantan Institute, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Dhêmerson Souza de Lima
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - João Paulo Mesquita Luiz
- Department of Pharmacology and Department of Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | - José Carlos Farias Alves-Filho
- Department of Pharmacology and Department of Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Alessandra Pontillo
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Karina Ramalho Bortoluci
- Department of Biological Sciences and Center for Cellular and Molecular Therapy (CTC-Mol),Federal University of São Paulo, São Paulo, Brazil
| | - Eliana L Faquim-Mauro
- Laboratory of Immunopathology, Butantan Institute, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil
| |
Collapse
|
47
|
Fraga-Silva TFDC, Maruyama SR, Sorgi CA, Russo EMDS, Fernandes APM, de Barros Cardoso CR, Faccioli LH, Dias-Baruffi M, Bonato VLD. COVID-19: Integrating the Complexity of Systemic and Pulmonary Immunopathology to Identify Biomarkers for Different Outcomes. Front Immunol 2021; 11:599736. [PMID: 33584667 PMCID: PMC7878380 DOI: 10.3389/fimmu.2020.599736] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
In the last few months, the coronavirus disease 2019 (COVID-19) pandemic has affected millions of people worldwide and has provoked an exceptional effort from the scientific community to understand the disease. Clinical evidence suggests that severe COVID-19 is associated with both dysregulation of damage tolerance caused by pulmonary immunopathology and high viral load. In this review article, we describe and discuss clinical studies that show advances in the understanding of mild and severe illness and we highlight major points that are critical for improving the comprehension of different clinical outcomes. The understanding of pulmonary immunopathology will contribute to the identification of biomarkers in an attempt to classify mild, moderate, severe and critical COVID-19 illness. The interface of pulmonary immunopathology and the identification of biomarkers are critical for the development of new therapeutic strategies aimed to reduce the systemic and pulmonary hyperinflammation in severe COVID-19.
Collapse
Affiliation(s)
- Thais Fernanda de Campos Fraga-Silva
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Sandra Regina Maruyama
- Department of Genetics and Evolution, Federal University of Sao Carlos, Sao Carlos, Brazil
| | - Carlos Arterio Sorgi
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Elisa Maria de Sousa Russo
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ana Paula Morais Fernandes
- Department of General and Specialized Nursing, School of Nursing of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Cristina Ribeiro de Barros Cardoso
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Lucia Helena Faccioli
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Marcelo Dias-Baruffi
- Department of Clinical Analysis, Toxicological and Food Science Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Vânia Luiza Deperon Bonato
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
48
|
Bothrops lanceolatus snake (Fer-de-lance) venom triggers inflammatory mediators' storm in human blood. Arch Toxicol 2021; 95:1129-1138. [PMID: 33398417 DOI: 10.1007/s00204-020-02959-0] [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] [Received: 09/30/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022]
Abstract
Systemic increased inflammatory mediators' levels are a hallmark in a plethora of pathological conditions, including thrombotic diseases as the envenomation by Bothrops lanceolatus snake. Multiple organ infarctions, which are not prevented by anticoagulant therapy, are the main cause of death on this envenomation. However, the potential mechanisms involved in these systemic reactions are underexplored. This study aimed to explore the potential systemic events which could contribute to thrombotic reactions on the envenomation by B. lanceolatus in an ex vivo human whole-blood model. B. lanceolatus venom elicited an inflammatory reaction, which was characterized by a strong complement activation, since we detected high C3a, C4a and C5a anaphylatoxins levels. Besides, the venom promoted soluble Terminal Complement Complex (sTCC) assembly. Complement activation was accompanied by intense lipid mediators' release, which included LTB4, PGE2 and TXB2. In addition, in the blood exposed to B. lanceolatus venom, we detected IL-1β, IL-6 and TNF-α interleukins production. Chemokines, including CCL2, CCL5 and CXCL8 were upregulated in the venom presence. These outcomes show that B. lanceolatus venom causes a strong inflammatory reaction in the blood favoring a potential setting to thrombi formation. Thus, inhibiting inflammatory mediators or their receptors may help in the envenomed patients' management.
Collapse
|
49
|
Boda F, Banfai K, Garai K, Kovacs B, Almasi A, Scheffer D, Sinkler RL, Csonka R, Czompoly T, Kvell K. Effect of Bitis gabonica and Dendroaspis angusticeps snake venoms on apoptosis-related genes in human thymic epithelial cells. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200057. [PMID: 33402885 PMCID: PMC7745260 DOI: 10.1590/1678-9199-jvatitd-2020-0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background: Certain environmental toxins permanently damage the thymic epithelium, accelerate immune senescence and trigger secondary immune pathologies. However, the exact underlying cellular mechanisms and pathways of permanent immune intoxication remain unknown. The aim of the present study was to demonstrate gene expressional changes of apoptosis-related cellular pathways in human thymic epithelial cells following exposure to snake venom from Bitis gabonica and Dendroaspis angusticeps. Methods: Snake venoms were characterized by analytical methods including reversed phase high-performance liquid chromatography and sodium dodecyl sulphate-polyacrylamide gel electrophoresis, then applied on human thymic epithelial cells (1889c) for 24 h at 10 μg/mL (as used in previous TaqMan Array study). Gene expressional changes restricted to apoptosis were assayed by TaqMan Array (Human Apoptosis Plate). Results: The most prominent gene expressional changes were shown by CASP5 (≈ 2.5 million-fold, confirmed by dedicated quantitative polymerase chain reaction) and CARD9 (0.016-fold) for B. gabonica, and BIRC7 (6.46-fold) and CASP1 (0.30-fold) for D. angusticeps. Conclusion: The observed apoptotic environment suggests that pyroptosis may be the dominant pathway through which B. gabonica and D. angusticeps snake venoms trigger thymic epithelial apoptosis following envenomation.
Collapse
Affiliation(s)
- Francisc Boda
- Department F1, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
| | - Krisztina Banfai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary.,Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary.,Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary
| | - Bela Kovacs
- Department F1, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
| | - Attila Almasi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Pecs, Pecs, Hungary
| | - Dalma Scheffer
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Reka Lambertne Sinkler
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Robert Csonka
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Tamas Czompoly
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary.,Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary
| |
Collapse
|
50
|
Salina ACG, Brandt SL, Klopfenstein N, Blackman A, Bazzano JMR, Sá-Nunes A, Byers-Glosson N, Brodskyn C, Tavares NM, Da Silva IBS, Medeiros AI, Serezani CH. Leukotriene B 4 licenses inflammasome activation to enhance skin host defense. Proc Natl Acad Sci U S A 2020; 117:30619-30627. [PMID: 33184178 PMCID: PMC7720147 DOI: 10.1073/pnas.2002732117] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The initial production of inflammatory mediators dictates host defense as well as tissue injury. Inflammasome activation is a constituent of the inflammatory response by recognizing pathogen and host-derived products and eliciting the production of IL-1β and IL-18 in addition to inducing a type of inflammatory cell death termed "pyroptosis." Leukotriene B4 (LTB4) is a lipid mediator produced quickly (seconds to minutes) by phagocytes and induces chemotaxis, increases cytokine/chemokine production, and enhances antimicrobial effector functions. Whether LTB4 directly activates the inflammasome remains to be determined. Our data show that endogenously produced LTB4 is required for the expression of pro-IL-1β and enhances inflammasome assembly in vivo and in vitro. Furthermore, LTB4-mediated Bruton's tyrosine kinase (BTK) activation is required for inflammasome assembly in vivo as well for IL-1β-enhanced skin host defense. Together, these data unveil a new role for LTB4 in enhancing the expression and assembly of inflammasome components and suggest that while blocking LTB4 actions could be a promising therapeutic strategy to prevent inflammasome-mediated diseases, exogenous LTB4 can be used as an adjuvant to boost inflammasome-dependent host defense.
Collapse
Affiliation(s)
- Ana Carolina Guerta Salina
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Stephanie L Brandt
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-3082
| | - Nathan Klopfenstein
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Amondrea Blackman
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
| | | | - Anderson Sá-Nunes
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Nicole Byers-Glosson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-3082
| | - Claudia Brodskyn
- Oswaldo Cruz Foundation, Gonçalo Moniz Institute, FIOCRUZ, Salvador 40296-710, Brazil
| | | | | | - Alexandra I Medeiros
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil
| | - C Henrique Serezani
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232;
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|