1
|
Xin W, Pan Y, Wei W, Gerner ST, Huber S, Juenemann M, Butz M, Bähr M, Huttner HB, Doeppner TR. TGF-β1 Decreases Microglia-Mediated Neuroinflammation and Lipid Droplet Accumulation in an In Vitro Stroke Model. Int J Mol Sci 2023; 24:17329. [PMID: 38139158 PMCID: PMC10743979 DOI: 10.3390/ijms242417329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Hypoxia triggers reactive microglial inflammation and lipid droplet (LD) accumulation under stroke conditions, although the mutual interactions between these two processes are insufficiently understood. Hence, the involvement of transforming growth factor (TGF)-β1 in inflammation and LD accumulation in cultured microglia exposed to hypoxia were analyzed herein. Primary microglia were exposed to oxygen-glucose deprivation (OGD) injury and lipopolysaccharide (LPS) stimulation. For analyzing the role of TGF-β1 patterns under such conditions, a TGF-β1 siRNA and an exogenous recombinant TGF-β1 protein were employed. Further studies applied Triacsin C, an inhibitor of LD formation, in order to directly assess the impact of LD formation on the modulation of inflammation. To assess mutual microglia-to-neuron interactions, a co-culture model of these cells was established. Upon OGD exposure, microglial TGF-β1 levels were significantly increased, whereas LPS stimulation yielded decreased levels. Elevating TGF-β1 expression proved highly effective in suppressing inflammation and reducing LD accumulation in microglia exposed to LPS. Conversely, inhibition of TGF-β1 led to the promotion of microglial cell inflammation and an increase in LD accumulation in microglia exposed to OGD. Employing the LD formation inhibitor Triacsin C, in turn, polarized microglia towards an anti-inflammatory phenotype. Such modulation of both microglial TGF-β1 and LD levels significantly affected the resistance of co-cultured neurons. This study provides novel insights by demonstrating that TGF-β1 plays a protective role against microglia-mediated neuroinflammation through the suppression of LD accumulation. These findings offer a fresh perspective on stroke treatment, suggesting the potential of targeting this pathway for therapeutic interventions.
Collapse
Affiliation(s)
- Wenqiang Xin
- Department of Neurology, University of Göttingen Medical School, 37075 Goettingen, Germany; (W.X.); (Y.P.); (W.W.); (M.B.)
| | - Yongli Pan
- Department of Neurology, University of Göttingen Medical School, 37075 Goettingen, Germany; (W.X.); (Y.P.); (W.W.); (M.B.)
| | - Wei Wei
- Department of Neurology, University of Göttingen Medical School, 37075 Goettingen, Germany; (W.X.); (Y.P.); (W.W.); (M.B.)
| | - Stefan T. Gerner
- Department of Neurology, University of Giessen Medical School, 35392 Giessen, Germany; (S.T.G.); (M.J.); (M.B.); (H.B.H.)
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University, 35032 Giessen, Germany
| | - Sabine Huber
- Department of Neurology, University of Giessen Medical School, 35392 Giessen, Germany; (S.T.G.); (M.J.); (M.B.); (H.B.H.)
| | - Martin Juenemann
- Department of Neurology, University of Giessen Medical School, 35392 Giessen, Germany; (S.T.G.); (M.J.); (M.B.); (H.B.H.)
| | - Marius Butz
- Department of Neurology, University of Giessen Medical School, 35392 Giessen, Germany; (S.T.G.); (M.J.); (M.B.); (H.B.H.)
- Heart and Brain Research Group, Kerckhoff Heart and Thorax Center, 61231 Bad Nauheim, Germany
| | - Mathias Bähr
- Department of Neurology, University of Göttingen Medical School, 37075 Goettingen, Germany; (W.X.); (Y.P.); (W.W.); (M.B.)
| | - Hagen B. Huttner
- Department of Neurology, University of Giessen Medical School, 35392 Giessen, Germany; (S.T.G.); (M.J.); (M.B.); (H.B.H.)
| | - Thorsten R. Doeppner
- Department of Neurology, University of Göttingen Medical School, 37075 Goettingen, Germany; (W.X.); (Y.P.); (W.W.); (M.B.)
- Department of Neurology, University of Giessen Medical School, 35392 Giessen, Germany; (S.T.G.); (M.J.); (M.B.); (H.B.H.)
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University, 35032 Giessen, Germany
- Department of Anatomy and Cell Biology, Medical University of Varna, 9238 Varna, Bulgaria
- Research Institute for Health Sciences and Technologies (SABITA), Medipol University, 100098 Istanbul, Turkey
| |
Collapse
|
2
|
Lemos ASDO, Campos LM, Granato JDT, Goliatt PVZC, Dib PRB, Hottz ED, Glanzmann N, Campos LC, Bizarro HDS, Chedier LM, Coimbra ES, Fabri RL. Mitracarpus frigidus reduces lipid metabolism and PGE2 levels in inflammatory cells. J Pharm Pharmacol 2023; 75:1388-1393. [PMID: 37487573 DOI: 10.1093/jpp/rgad069] [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: 02/13/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023]
Abstract
OBJECTIVES To evaluate the ability of the aqueous extract of Mitracarpus frigidus (MFAq) to inhibit lipid body formation and inflammatory mediator production in macrophages stimulated with lipopolysaccharide (LPS) and interferon gamma (IFN-γ). METHODS MFAq was chemically characterized by ultrafast liquid chromatography/quadruple time-of-flight tandem mass spectrometry. The macrophages obtained from mice were incubated with MFAq. Cell viability and membrane integrity were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and propidium iodide assays, respectively. Moreover, NO, reactive oxygen species (ROS), transforming growth factor beta (TGF-β), prostaglandin E2 (PGE2) levels and lipid bodies (LBs) were examined in macrophages that were stimulated with LPS and IFN-γ and treated with MFAq. Finally, molecular docking analysis was conducted to investigate the interaction of MFAq with the cyclooxygenase 2 (COX-2) enzyme. KEY FINDINGS Chlorogenic acid, clarinoside, harounoside, rutin, kaempferol-3O-rutinoside and 2-azaanthraquinone were identified in MFAq. MFAq significantly inhibited NO, ROS and LBs, and did not affect the membrane integrity of macrophages. MFAq-treated cells showed significantly lower levels of TGF-β and PGE2. Molecular docking demonstrated that the compounds found in MFAq are able to inhibit COX-2 by binding to important residues in the catalytic site. CONCLUSIONS MFAq interferes with lipid metabolism in stimulated macrophages, leading to the reduction of important inflammatory mediators. Furthermore, MFAq can directly inhibit the COX-2 enzyme or inhibit its expression owing to its ability to reduce NO production.
Collapse
Affiliation(s)
- Ari Sérgio de O Lemos
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Lara M Campos
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Juliana da T Granato
- Department of Parasitology, Microbiology, and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Priscila V Z C Goliatt
- Department of Computer Science, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Paula R B Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Nícolas Glanzmann
- Department of Chemistry, Institute of Exact Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laíris C Campos
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Heloisa D S Bizarro
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Luciana M Chedier
- Plant Chemistry Laboratory, Department of Botany, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Elaine S Coimbra
- Department of Parasitology, Microbiology, and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Rodrigo L Fabri
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
3
|
Hammoudeh N, Soukkarieh C, Murphy DJ, Hanano A. Mammalian lipid droplets: structural, pathological, immunological and anti-toxicological roles. Prog Lipid Res 2023; 91:101233. [PMID: 37156444 DOI: 10.1016/j.plipres.2023.101233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Mammalian lipid droplets (LDs) are specialized cytosolic organelles consisting of a neutral lipid core surrounded by a membrane made up of a phospholipid monolayer and a specific population of proteins that varies according to the location and function of each LD. Over the past decade, there have been significant advances in the understanding of LD biogenesis and functions. LDs are now recognized as dynamic organelles that participate in many aspects of cellular homeostasis plus other vital functions. LD biogenesis is a complex, highly-regulated process with assembly occurring on the endoplasmic reticulum although aspects of the underpinning molecular mechanisms remain elusive. For example, it is unclear how many enzymes participate in the biosynthesis of the neutral lipid components of LDs and how this process is coordinated in response to different metabolic cues to promote or suppress LD formation and turnover. In addition to enzymes involved in the biosynthesis of neutral lipids, various scaffolding proteins play roles in coordinating LD formation. Despite their lack of ultrastructural diversity, LDs in different mammalian cell types are involved in a wide range of biological functions. These include roles in membrane homeostasis, regulation of hypoxia, neoplastic inflammatory responses, cellular oxidative status, lipid peroxidation, and protection against potentially toxic intracellular fatty acids and lipophilic xenobiotics. Herein, the roles of mammalian LDs and their associated proteins are reviewed with a particular focus on their roles in pathological, immunological and anti-toxicological processes.
Collapse
Affiliation(s)
- Nour Hammoudeh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Denis J Murphy
- School of Applied Sciences, University of South Wales, Pontypridd, CF37 1DL, Wales, United Kingdom..
| | - Abdulsamie Hanano
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria (AECS), P.O. Box 6091, Damascus, Syria..
| |
Collapse
|
4
|
Rosa IF, Peçanha APB, Carvalho TRB, Alexandre LS, Ferreira VG, Doretto LB, Souza BM, Nakajima RT, da Silva P, Barbosa AP, Gomes-de-Pontes L, Bomfim CG, Machado-Santelli GM, Condino-Neto A, Guzzo CR, Peron JPS, Andrade-Silva M, Câmara NOS, Garnique AMB, Medeiros RJ, Ferraris FK, Barcellos LJG, Correia-Junior JD, Galindo-Villegas J, Machado MFR, Castoldi A, Oliveira SL, Costa CC, Belo MAA, Galdino G, Sgro GG, Bueno NF, Eto SF, Veras FP, Fernandes BHV, Sanches PRS, Cilli EM, Malafaia G, Nóbrega RH, Garcez AS, Carrilho E, Charlie-Silva I. Photobiomodulation Reduces the Cytokine Storm Syndrome Associated with COVID-19 in the Zebrafish Model. Int J Mol Sci 2023; 24:ijms24076104. [PMID: 37047078 PMCID: PMC10094635 DOI: 10.3390/ijms24076104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
Although the exact mechanism of the pathogenesis of coronavirus SARS-CoV-2 (COVID-19) is not fully understood, oxidative stress and the release of pro-inflammatory cytokines have been highlighted as playing a vital role in the pathogenesis of the disease. In this sense, alternative treatments are needed to reduce the level of inflammation caused by COVID-19. Therefore, this study aimed to investigate the potential effect of red photobiomodulation (PBM) as an attractive therapy to downregulate the cytokine storm caused by COVID-19 in a zebrafish model. RT-qPCR analyses and protein-protein interaction prediction among SARS-CoV-2 and Danio rerio proteins showed that recombinant Spike protein (rSpike) was responsible for generating systemic inflammatory processes with significantly increased levels of pro-inflammatory (il1b, il6, tnfa, and nfkbiab), oxidative stress (romo1) and energy metabolism (slc2a1a and coa1) mRNA markers, with a pattern similar to those observed in COVID-19 cases in humans. On the other hand, PBM treatment was able to decrease the mRNA levels of these pro-inflammatory and oxidative stress markers compared with rSpike in various tissues, promoting an anti-inflammatory response. Conversely, PBM promotes cellular and tissue repair of injured tissues and significantly increases the survival rate of rSpike-inoculated individuals. Additionally, metabolomics analysis showed that the most-impacted metabolic pathways between PBM and the rSpike treated groups were related to steroid metabolism, immune system, and lipid metabolism. Together, our findings suggest that the inflammatory process is an incisive feature of COVID-19 and red PBM can be used as a novel therapeutic agent for COVID-19 by regulating the inflammatory response. Nevertheless, the need for more clinical trials remains, and there is a significant gap to overcome before clinical trials can commence.
Collapse
Affiliation(s)
- Ivana F Rosa
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 01049-010, Brazil
| | - Ana P B Peçanha
- Department of Orthodontics, São Leopoldo Mandic College, Campinas 13045-755, Brazil
| | - Tábata R B Carvalho
- Department of Orthodontics, São Leopoldo Mandic College, Campinas 13045-755, Brazil
| | - Leonardo S Alexandre
- Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
- The National Institute of Science and Technology in Bioanalyses, INCTBio, Campinas 13083-970, Brazil
| | - Vinícius G Ferreira
- Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
- The National Institute of Science and Technology in Bioanalyses, INCTBio, Campinas 13083-970, Brazil
| | - Lucas B Doretto
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 01049-010, Brazil
| | - Beatriz M Souza
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 01049-010, Brazil
| | - Rafael T Nakajima
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 01049-010, Brazil
| | - Patrick da Silva
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Ana P Barbosa
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Leticia Gomes-de-Pontes
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Camila G Bomfim
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | | | - Antonio Condino-Neto
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Cristiane R Guzzo
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Jean P S Peron
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Magaiver Andrade-Silva
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Niels O S Câmara
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | - Anali M B Garnique
- Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-220, Brazil
| | | | | | - Leonardo J G Barcellos
- Laboratório de Fisiologia de Peixes, Programa de Pós-Graduação em Bioexperimentação, Escola de Ciências Agrárias, Inovação e Negócios, Universidade de Passo Fundo, Passo Fundo 99052-900, Brazil
| | - Jose D Correia-Junior
- Institute of Biomedical Sciences, Federal University Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Jorge Galindo-Villegas
- Department of Genomics, Faculty of Biosciences and Aquaculture, Nord University, 8026 Bodø, Norway
| | - Mônica F R Machado
- Biological Sciences Special Academic Unit, Federal University of Jatai, Jatai 75804-020, Brazil
| | - Angela Castoldi
- Keizo Asami Institute, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Susana L Oliveira
- School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, Brazil
| | - Camila C Costa
- School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, Brazil
| | - Marco A A Belo
- School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, Brazil
| | - Giovane Galdino
- Institute of Motricity Sciences, Department of Physical Therapy, Federal University of Alfenas, Alfenas 37133-840, Brazil
| | - Germán G Sgro
- Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo 14040-900, Brazil
| | - Natalia F Bueno
- Integrated Structural Biology Platform, Carlos Chagas Institute, FIOCRUZ Paraná, Curitiba 81310-020, Brazil
| | - Silas F Eto
- Center of Innovation and Development, Laboratory of Development and Innovation Butantan Institute, São Paulo 69310-000, Brazil
| | - Flávio P Veras
- Faculty of Medicine, University of São Paulo (USP), Ribeirão Preto 14040-900, Brazil
| | - Bianca H V Fernandes
- Laboratory of Genetic and Sanitary Control, Technical Board of Support for Teaching and Research, Faculty of Medicine, University of Sao Paulo, São Paulo 01246-903, Brazil
| | - Paulo R S Sanches
- Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-060, Brazil
| | - Eduardo M Cilli
- Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-060, Brazil
| | - Guilherme Malafaia
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute, Urutaí Campus, Urutaí 75790-000, Brazil
| | - Rafael H Nóbrega
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 01049-010, Brazil
| | - Aguinaldo S Garcez
- Department of Orthodontics, São Leopoldo Mandic College, Campinas 13045-755, Brazil
| | - Emanuel Carrilho
- Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
- The National Institute of Science and Technology in Bioanalyses, INCTBio, Campinas 13083-970, Brazil
| | - Ives Charlie-Silva
- Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-060, Brazil
| |
Collapse
|
5
|
Lopes JA, Boeno CN, Paloschi MV, Silva MDS, Rego CMA, Pires WL, Santana HM, Chaves YO, Rodrigues MMDS, Lima AM, Setúbal SDS, Soares AM, Zuliani JP. Phenotypic, functional and plasticity features of human PBMCs induced by venom secreted PLA 2s. Mol Immunol 2023; 155:135-152. [PMID: 36812762 DOI: 10.1016/j.molimm.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023]
Abstract
Bothrops venom contains a high amount of secreted phospholipase A2 (sPLA2s) enzymes responsible for the inflammatory reaction and activation of leukocytes in cases of envenoming. PLA2s are proteins that have enzymatic activity and can hydrolyze phospholipids at the sn-2 position, thereby releasing fatty acids and lysophospholipids precursors of eicosanoids, which are significant mediators of inflammatory conditions. Whether these enzymes have a role in the activation and function of peripheral blood mononuclear cells (PBMCs) is not known. Here we show for the first time how two secreted PLA2s (BthTX-I and BthTX-II) isolated from the venom of Bothrops jararacussu affect the function and polarization of PBMCs. Neither BthTX-I nor BthTX-II exhibited significant cytotoxicity to isolated PBMCs compared with the control at any of the time points studied. RT-qPCR and enzyme-linked immunosorbent assays were used to determine changes in gene expression and the release of pro-inflammatory (TNF-α, IL-6, and IL-12) and anti-inflammatory (TGF-β and IL-10) cytokines, respectively, during the cell differentiation process. Lipid droplets formation and phagocytosis were also investigated. Monocytes/macrophages were labeled with anti-CD14, -CD163, and -CD206 antibodies to assay cell polarization. Both toxins caused a heterogeneous morphology (M1 and M2) on days 1 and 7 based on immunofluorescence analysis, revealing the considerable flexibility of these cells even in the presence of typical polarization stimuli. Thus, these findings indicate that the two sPLA2s trigger both immune response profiles in PBMCs indicating a significant degree of cell plasticity, which may be crucial for understanding the consequences of snake envenoming.
Collapse
Affiliation(s)
- Jéssica Amaral Lopes
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Charles Nunes Boeno
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Mauro Valentino Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Milena Daniela Souza Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Cristina Matiele Alves Rego
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Weverson Luciano Pires
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Hallison Mota Santana
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Yury Oliveira Chaves
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Fundação Oswaldo Cruz, FIOCRUZ Amazônia, Manaus, AM, Brazil
| | - Moreno Magalhães de Souza Rodrigues
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Análise e Visualização de Dados, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Anderson M Lima
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Brazil
| | - Sulamita da S Setúbal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Andreimar M Soares
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Brazil
| | - Juliana P Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Brazil.
| |
Collapse
|
6
|
Shrimp Lipid Droplet Protein Perilipin Involves in the Pathogenesis of AHPND-Causing Vibrio parahaemolyticus. Int J Mol Sci 2022; 23:ijms231810520. [PMID: 36142431 PMCID: PMC9501514 DOI: 10.3390/ijms231810520] [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: 08/12/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Acute hepatopancreatic necrosis disease (AHPND), caused by a unique strain of Vibrio parahaemolyticus (Vp (AHPND)), has become the world’s most severe debilitating disease in cultured shrimp. Thus far, the pathogenesis of AHPND remains largely unknow. Herein, in Litopenaeus vannamei, we found that a Vp (AHPND) infection significantly increased the expression of lipid droplets (LDs) protein LvPerilipin, as well as promoted the formation of LDs. In addition, the knockdown of LvPerilipin increased the shrimp survival rate in response to the Vp (AHPND) infection, and inhibited the proliferation of Vp (AHPND). Furthermore, we demonstrated that LvPerilipin depletion could increase the production of reactive oxygen species (ROS), which may be responsible for the decreased Vp (AHPND) proliferation. Taken together, our current data for the first time reveal that the shrimp lipid droplets protein Perilipin is involved in the pathogenesis of Vp (AHPND) via promoting LDs accumulation and decreasing ROS production.
Collapse
|
7
|
Triglyceride breakdown from lipid droplets regulates the inflammatory response in macrophages. Proc Natl Acad Sci U S A 2022; 119:e2114739119. [PMID: 35302892 PMCID: PMC8944848 DOI: 10.1073/pnas.2114739119] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lipid droplets (LDs) are ubiquitous organelles that play important roles in cellular energy homeostasis, tightly regulating the accumulation and release of lipids. In macrophages, lipids accumulate in LDs during inflammation. However, it is unclear how inflammatory activation promotes the accumulation of lipids in LDs, and how the dynamic between lipid accumulation and breakdown could drive or inhibit inflammation. Elucidating the role of lipid accumulation during inflammation may provide important knowledge to influence inflammatory processes during health and disease. We identify the importance of the hypoxia-inducible lipid droplet–associated protein and the intracellular adipose triglyceride lipase in the regulation of lipid accumulation and breakdown in inflammatory macrophages. Furthermore, we determine the regulatory effect of lipid breakdown from LDs in supporting inflammation. In response to inflammatory activation by pathogens, macrophages accumulate triglycerides in intracellular lipid droplets. The mechanisms underlying triglyceride accumulation and its exact role in the inflammatory response of macrophages are not fully understood. Here, we aim to further elucidate the mechanism and function of triglyceride accumulation in the inflammatory response of activated macrophages. Lipopolysaccharide (LPS)-mediated activation markedly increased triglyceride accumulation in macrophages. This increase could be attributed to up-regulation of the hypoxia-inducible lipid droplet–associated (HILPDA) protein, which down-regulated adipose triglyceride lipase (ATGL) protein levels, in turn leading to decreased ATGL-mediated triglyceride hydrolysis. The reduction in ATGL-mediated lipolysis attenuated the inflammatory response in macrophages after ex vivo and in vitro activation, and was accompanied by decreased production of prostaglandin-E2 (PGE2) and interleukin-6 (IL-6). Overall, we provide evidence that LPS-mediated activation of macrophages suppresses lipolysis via induction of HILPDA, thereby reducing the availability of proinflammatory lipid precursors and suppressing the production of PGE2 and IL-6.
Collapse
|
8
|
Wang X, Chi Y, Li J, Pang Y, Li Q. Morphological characteristics and a single-cell analysis provide insights into function of immune and fat storage in the lamprey supraneural body. Int J Biochem Cell Biol 2022; 142:106131. [PMID: 34838690 DOI: 10.1016/j.biocel.2021.106131] [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: 06/30/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 10/19/2022]
Abstract
The supraneural body, also known as dorsal fat body is considered from adipose progenitors, and possesses hematopoietic activity. However, in-depth knowledge of cell-type by single-cell transcriptome sequencing and physiological functions are still lacking. Here, we determined at least four types of cells, such as white adipocytes, granulocytes, lymphocytes, and red blood cells by using 10 ×Genomics single-cell RNA sequencing (scRNA-Seq), hematoxylin-eosin (HE) staining, electron microscopy, immunofluorescence, and histochemistry. Additionally, most immune cells contain scattered small fat droplets except for white adipocytes with one large lipid droplet. The content of triglyceride in supraneural body is the highest compared with other tissues. The mRNA expression of both lipolysis-related genes and brown adipocytes-specific marker genes were up-regulated in supraneural body cells in response to epinephrine. Taken together, these data indicate that the supraneural body may play an important role in immune and fat storage. Our findings not only provided detailed insights into the unique molecular make-up of the supraneural body tissue, but also shed new light on future analyses of physiological functions in immune or lipid regulating.
Collapse
Affiliation(s)
- Xiaotong Wang
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| | - Yan Chi
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| | - Jun Li
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| | - Yue Pang
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian 116081, China.
| | - Qingwei Li
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian 116081, China.
| |
Collapse
|
9
|
Elghoul M, El-Gendy SAA, Alsafy MAM. Ultrastructural investigation of the pneumocytes in piglets that live in a trashed environment. Morphologie 2021; 106:294-299. [PMID: 34933787 DOI: 10.1016/j.morpho.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 11/15/2022]
Abstract
The present study provides the ultrastructure of the pneumocytes types II and I in piglets living in the trash environment. Samples of the lungs of twelve piglets two months old were used. By light microscopy, the pneumocytes type I were squamous and somewhat flattened cells, while pneumocytes type II was cuboidal to spherical projected within the alveolar lumen and characterized by a spherical nucleus and foamy cytoplasm, it contained vacuolated bodies that were polygonal with variable size manly at the poles of the cell. The electron microscopy investigation showed blood air barrier between the endothelial lining of pneumocytes type I and therefore the endothelial lining of blood capillary and their nucleus were irregular in shape varied from nearly irregular triangular to polygon rough endoplasmic reticulum represented at their cytoplasm. The pneumocytes type II were frequently covered by pneumocytes type I extensions and united to them by a tight junction. It had been characterized by a high number of mitochondria within the cytoplasm and vacuolated bodies encircled the nucleus and at the two extremities of the cell. The lamellar vacuolated bodies were connected to the endoplasmic reticulum membranes and therefore the intravascular macrophages were attached to the endothelial cells within the pulmonary capillaries until two months old piglets. The occurrence of the intravascular macrophages could be attributed to the higher resistance to the respiratory diseases of the piglets.
Collapse
Affiliation(s)
- M Elghoul
- Histology and Cytology Department, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt.
| | - S A A El-Gendy
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt.
| | - M A M Alsafy
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
10
|
Kiarely Souza E, Pereira-Dutra FS, Rajão MA, Ferraro-Moreira F, Goltara-Gomes TC, Cunha-Fernandes T, Santos JDC, Prestes EB, Andrade WA, Zamboni DS, Bozza MT, Bozza PT. Lipid droplet accumulation occurs early following Salmonella infection and contributes to intracellular bacterial survival and replication. Mol Microbiol 2021; 117:293-306. [PMID: 34783412 DOI: 10.1111/mmi.14844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 01/20/2023]
Abstract
Salmonellosis is a public health problem caused by Salmonella sp., a highly adapted facultative intracellular pathogen. After internalization, Salmonella sp. Manipulates several host processes, mainly through the activation of the type III secretion system (T3SS), including modification of host lipid metabolism and lipid droplet (LD) accumulation. LDs are dynamic and complex lipid-rich organelles involved in several cellular processes. The present study investigated the mechanism involved in LD biogenesis in Salmonella-infected macrophages and its role in bacterial pathogenicity. Here, we reported that S. Typhimurium induced a rapid time-dependent increase of LD formation in macrophages. The LD biogenesis was demonstrated to depend on Salmonella's viability and SPI1-related T3SS activity, with the participation of Toll-Like Receptor (TLR) signaling. We also observed that LD accumulation occurs through TLR2-dependent signaling and is counter-regulated by TLR4. Last, the pharmacologic modulation of LD formation by inhibiting diacylglycerol O-acyltransferase 1 (DGAT1) and cytosolic phospholipase A2 (cPLA2) significantly reduced the intracellular bacterial proliferation and impaired the prostaglandin E2 (PGE2 ) synthesis. Collectively, our data suggest the role of LDs on S. typhimurium intracellular survival and replication in macrophages. This data set provides new perspectives for future investigations about LDs in host-pathogen interaction.
Collapse
Affiliation(s)
- Ellen Kiarely Souza
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Program of Immunology and Inflammation, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Filipe S Pereira-Dutra
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Matheus A Rajão
- Program of Immunology and Tumor Biology, Instituto Nacional do Câncer, INCA, Rio de Janeiro, Brazil
| | - Felipe Ferraro-Moreira
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Taynná C Goltara-Gomes
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Tamires Cunha-Fernandes
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Julia da Cunha Santos
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Elisa B Prestes
- Laboratory of Inflammation and Immunity, Department of Immunity, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Warrison A Andrade
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Dario S Zamboni
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo T Bozza
- Laboratory of Inflammation and Immunity, Department of Immunity, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Wang L, Lin J, Yu J, Yang K, Sun L, Tang H, Pan L. Downregulation of Perilipin1 by the Immune Deficiency Pathway Leads to Lipid Droplet Reconfiguration and Adaptation to Bacterial Infection in Drosophila. THE JOURNAL OF IMMUNOLOGY 2021; 207:2347-2358. [PMID: 34588219 DOI: 10.4049/jimmunol.2100343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
Lipid droplets (LDs), the highly dynamic intracellular organelles, are critical for lipid metabolism. Dynamic alterations in the configurations and functions of LDs during innate immune responses to bacterial infections and the underlying mechanisms, however, remain largely unknown. In this study, we trace the time-course morphology of LDs in fat bodies of Drosophila after transient bacterial infection. Detailed analysis shows that perilipin1 (plin1), a core gene involved in the regulation of LDs, is suppressed by the immune deficiency signaling, one major innate immune pathway in Drosophila During immune activation, downregulated plin1 promotes the enlargement of LDs, which in turn alleviates immune reaction-associated reactive oxygen species stress. Thus, the growth of LDs is likely an active adaptation to maintain redox homeostasis in response to immune deficiency activation. Therefore, our study provides evidence that plin1 serves as a modulator on LDs' reconfiguration in regulating infection-induced pathogenesis, and plin1 might be a potential therapeutic target for coordinating inflammation resolution and lipid metabolism.
Collapse
Affiliation(s)
- Lei Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.,The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiaxin Lin
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Junjing Yu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China; and
| | - Kaiyan Yang
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Li Sun
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Hong Tang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China;
| | - Lei Pan
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China; .,University of Chinese Academy of Sciences, Beijing, China.,Chinese Academy of Sciences Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Pereira-Dutra FS, Bozza PT. Lipid droplets diversity and functions in inflammation and immune response. Expert Rev Proteomics 2021; 18:809-825. [PMID: 34668810 DOI: 10.1080/14789450.2021.1995356] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Lipid droplets (LDs) are dynamic and evolutionary conserved lipid-enriched organelles composed of a core of neutral lipids surrounded by a monolayer of phospholipids associated with a diverse array of proteins that are cell- and stimulus-regulated. Far beyond being simply a deposit of neutral lipids, accumulating evidence demonstrate that LDs act as spatial and temporal local for lipid and protein compartmentalization and signaling organization. AREAS COVERED This review focuses on the progress in our understanding of LD protein diversity and LD functions in the context of cell signaling and immune responses, highlighting the relationship between LD composition with the multiple roles of this organelle in immunometabolism, inflammation and host-response to infection. EXPERT OPINION LDs are essential platforms for various cellular processes, including metabolic regulation, cell signaling, and immune responses. The functions of LD in infection and inflammatory disease are associated with the dynamic and complexity of their proteome. Our contemporary view place LDs as critical regulators of different inflammatory and infectious diseases and key markers of leukocyte activation.
Collapse
Affiliation(s)
- Filipe S Pereira-Dutra
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Monson EA, Trenerry AM, Laws JL, Mackenzie JM, Helbig KJ. Lipid droplets and lipid mediators in viral infection and immunity. FEMS Microbiol Rev 2021; 45:fuaa066. [PMID: 33512504 PMCID: PMC8371277 DOI: 10.1093/femsre/fuaa066] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Lipid droplets (LDs) contribute to key pathways important for the physiology and pathophysiology of cells. In a homeostatic view, LDs regulate the storage of neutral lipids, protein sequestration, removal of toxic lipids and cellular communication; however, recent advancements in the field show these organelles as essential for various cellular stress response mechanisms, including inflammation and immunity, with LDs acting as hubs that integrate metabolic and inflammatory processes. The accumulation of LDs has become a hallmark of infection, and is often thought to be virally driven; however, recent evidence is pointing to a role for the upregulation of LDs in the production of a successful immune response to viral infection. The fatty acids housed in LDs are also gaining interest due to the role that these lipid species play during viral infection, and their link to the synthesis of bioactive lipid mediators that have been found to have a very complex role in viral infection. This review explores the role of LDs and their subsequent lipid mediators during viral infections and poses a paradigm shift in thinking in the field, whereby LDs may play pivotal roles in protecting the host against viral infection.
Collapse
Affiliation(s)
- Ebony A Monson
- School of Life Sciences, La Trobe University, Melbourne, Australia, 3083
| | - Alice M Trenerry
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia, 3000
| | - Jay L Laws
- School of Life Sciences, La Trobe University, Melbourne, Australia, 3083
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia, 3000
| | - Karla J Helbig
- School of Life Sciences, La Trobe University, Melbourne, Australia, 3083
| |
Collapse
|
14
|
de Lira MN, Bolini L, Amorim NRT, Silva-Souza HA, Diaz BL, Canetti C, Persechini PM, Bandeira-Melo C. Acute catabolism of leukocyte lipid bodies: Characterization of a nordihydroguaiaretic acid (NDGA)-induced proteasomal-dependent model. Prostaglandins Leukot Essent Fatty Acids 2021; 171:102320. [PMID: 34303171 DOI: 10.1016/j.plefa.2021.102320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Cytoplasmic availability of leukocyte lipid bodies is controlled by a highly regulated cycle of opposing biogenesis- and catabolism-related events. While leukocyte biogenic machinery is well-characterized, lipid body catabolic mechanisms are yet mostly unknown. Here, we demonstrated that nordihydroguaiaretic acid (NDGA) very rapidly decreases the numbers of pre-formed lipid bodies within lipid body-enriched cytoplasm of mouse leukocytes - macrophages, neutrophils and eosinophils. NDGA mechanisms driving leukocyte lipid body disappearance were not related to loss of cell viability, 5-lipoxygenase inhibition, ATP autocrine/paracrine activity, or biogenesis inhibition. Proteasomal-dependent breakdown of lipid bodies appears to control NDGA-driven leukocyte lipid body reduction, since it was Bortezomib-sensitive in macrophages, neutrophils and eosinophils. Our findings unveil an acute NDGA-triggered lipid body catabolic event - a novel experimental model for the still neglected research area on leukocyte lipid body catabolism, additionally favoring further insights on proteasomal contribution to lipid body breakdown.
Collapse
Affiliation(s)
- Maria N de Lira
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Laboratório de Imunobiofisica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; BioMed X Institute (GmbH), Heidelberg, Germany
| | - Lukas Bolini
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natália R T Amorim
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hercules A Silva-Souza
- Laboratório de Imunobiofisica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Divisão de Verificação e Estudos Técnico-Científicos, Instituto Nacional de Metrologia Qualidade e Tecnologia, Duque de Caxias Rio de Janeiro, Brazil
| | - Bruno L Diaz
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudio Canetti
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro M Persechini
- Laboratório de Imunobiofisica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; This paper is dedicated to the memory of our dear colleague and friend Pedro M. Persechini, who passed prematurely and whose devotion to understanding the mechanisms of action of NDGA was unsurpassed
| | - Christianne Bandeira-Melo
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Intracellular lipid droplet accumulation occurs early following viral infection and is required for an efficient interferon response. Nat Commun 2021; 12:4303. [PMID: 34262037 PMCID: PMC8280141 DOI: 10.1038/s41467-021-24632-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/25/2021] [Indexed: 02/06/2023] Open
Abstract
Lipid droplets (LDs) are increasingly recognized as critical organelles in signalling events, transient protein sequestration and inter-organelle interactions. However, the role LDs play in antiviral innate immune pathways remains unknown. Here we demonstrate that induction of LDs occurs as early as 2 h post-viral infection, is transient and returns to basal levels by 72 h. This phenomenon occurs following viral infections, both in vitro and in vivo. Virally driven in vitro LD induction is type-I interferon (IFN) independent, and dependent on Epidermal Growth Factor Receptor (EGFR) engagement, offering an alternate mechanism of LD induction in comparison to our traditional understanding of their biogenesis. Additionally, LD induction corresponds with enhanced cellular type-I and -III IFN production in infected cells, with enhanced LD accumulation decreasing viral replication of both Herpes Simplex virus 1 (HSV-1) and Zika virus (ZIKV). Here, we demonstrate, that LDs play vital roles in facilitating the magnitude of the early antiviral immune response specifically through the enhanced modulation of IFN following viral infection, and control of viral replication. By identifying LDs as a critical signalling organelle, this data represents a paradigm shift in our understanding of the molecular mechanisms which coordinate an effective antiviral response.
Collapse
|
16
|
Brink JTR, Fourie R, Sebolai O, Albertyn J, Pohl CH. The role of lipid droplets in microbial pathogenesis. J Med Microbiol 2021; 70. [PMID: 34184983 DOI: 10.1099/jmm.0.001383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The nonpolar lipids present in cells are mainly triacylglycerols and steryl esters. When cells are provided with an abundance of nutrients, these storage lipids accumulate. As large quantities of nonpolar lipids cannot be integrated into membranes, they are isolated from the cytosolic environment in lipid droplets. As specialized, inducible cytoplasmic organelles, lipid droplets have functions beyond the regulation of lipid metabolism, in cell signalling and activation, membrane trafficking and control of inflammatory mediator synthesis and secretion. Pathogens, including fungi, viruses, parasites, or intracellular bacteria can induce and may benefit from lipid droplets in infected cells. Here we review biogenesis of lipid droplets as well as the role of lipid droplets in the pathogenesis of selected viruses, bacteria, protists and yeasts.
Collapse
Affiliation(s)
- Jacobus T R Brink
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Ruan Fourie
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Olihile Sebolai
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Jacobus Albertyn
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Carolina H Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
17
|
Ralhan I, Chang CL, Lippincott-Schwartz J, Ioannou MS. Lipid droplets in the nervous system. J Cell Biol 2021; 220:e202102136. [PMID: 34152362 PMCID: PMC8222944 DOI: 10.1083/jcb.202102136] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 01/20/2023] Open
Abstract
Lipid droplets are dynamic intracellular lipid storage organelles that respond to the physiological state of cells. In addition to controlling cell metabolism, they play a protective role for many cellular stressors, including oxidative stress. Despite prior descriptions of lipid droplets appearing in the brain as early as a century ago, only recently has the role of lipid droplets in cells found in the brain begun to be understood. Lipid droplet functions have now been described for cells of the nervous system in the context of development, aging, and an increasing number of neuropathologies. Here, we review the basic mechanisms of lipid droplet formation, turnover, and function and discuss how these mechanisms enable lipid droplets to function in different cell types of the nervous system under healthy and pathological conditions.
Collapse
Affiliation(s)
- Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Chi-Lun Chang
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | | | - Maria S. Ioannou
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
18
|
Boucher DM, Vijithakumar V, Ouimet M. Lipid Droplets as Regulators of Metabolism and Immunity. IMMUNOMETABOLISM 2021; 3. [DOI: 10.20900/immunometab20210021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/28/2021] [Indexed: 01/03/2025]
Abstract
Abstract
A hallmark of sterile and nonsterile inflammation is the increased accumulation of cytoplasmic lipid droplets (LDs) in non-adipose cells. LDs are ubiquitous organelles specialized in neutral lipid storage and hydrolysis. Originating in the ER, LDs are comprised of a core of neutral lipids (cholesterol esters, triglycerides) surrounded by a phospholipid monolayer and several LD-associated proteins. The perilipin (PLIN1-5) family are the most abundant structural proteins present on the surface of LDs. While PLIN1 is primarily expressed in adipocytes, PLIN2 and PLIN3 are ubiquitously expressed. LDs also acquire a host of enzymes and proteins that regulate LD metabolism. Amongst these are neutral lipases and selective lipophagy factors that promote hydrolysis of LD-associated neutral lipid. In addition, LDs physically associate with other organelles such as mitochondria through inter-organelle membrane contact sites that facilitate lipid transport. Beyond serving as a source of energy storage, LDs participate in inflammatory and infectious diseases, regulating both innate and adaptive host immune responses. Here, we review recent studies on the role of LDs in the regulation of immunometabolism.
Collapse
Affiliation(s)
- Dominique M. Boucher
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Viyashini Vijithakumar
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Mireille Ouimet
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| |
Collapse
|
19
|
Wang H, Wen X, Yan M, Chang M, Zhang G, Peng W, Wu Y, Shen Y, Zhou J, Li H. The role of perilipin 2 in Pseudomonas aeruginosa pulmonary infection. Respir Physiol Neurobiol 2020; 281:103497. [DOI: 10.1016/j.resp.2020.103497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/23/2020] [Accepted: 07/14/2020] [Indexed: 01/24/2023]
|
20
|
Tang M, Hu Z, Rao C, Chen J, Yuan S, Zhang J, Mao C, Yan J, Xia Y, Zhang M, Yue J, Xiang Y, Xie J, Mao X, Li Q. Burkholderia pseudomallei interferes with host lipid metabolism via NR1D2-mediated PNPLA2/ATGL suppression to block autophagy-dependent inhibition of infection. Autophagy 2020; 17:1918-1933. [PMID: 32777979 PMCID: PMC8386627 DOI: 10.1080/15548627.2020.1801270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
Burkholderia pseudomallei: which causes melioidosis with high mortality in humans, has become a global public health concern. Recently, infection-driven lipid droplet accumulation has been related to the progression of host-pathogen interactions, and its contribution to the pathogenesis of infectious disease has been investigated. Here, we demonstrated that B. pseudomallei infection actively induced a time-dependent increase in the number and size of lipid droplets in human lung epithelial cells and macrophages. We also found that lipid droplet accumulation following B. pseudomallei infection was associated with downregulation of PNPLA2/ATGL (patatin like phospholipase domain containing 2) and lipophagy inhibition. Functionally, lipid droplet accumulation, facilitated via PNPLA2 downregulation, inhibited macroautophagic/autophagic flux and, thus, hindered autophagy-dependent inhibition of B. pseudomallei infection in lung epithelial cells. Mechanistically, we further revealed that nuclear receptor NR1D2 might be involved in the suppression of PNPLA2 after cell exposure to B. pseudomallei. Taken together, our findings unraveled an evolutionary strategy, by which B. pseudomallei interferes with the host lipid metabolism, to block autophagy-dependent suppression of infection. This study proposes potential targets for clinical therapy of melioidosis.Abbreviations: 3-MA: 3-methyladenine; ACTB: actin beta; ATG7: autophagy related 7; B. pseudomallei: Burkholderia pseudomallei; CFU: colony-forming unit; DG: diglyceride; FASN: fatty acid synthase; GFP: green fluorescent protein; LAMP1: lysosomal associated membrane protein 1; LC-MS/MS: liquid chromatography-tandem mass spectrometry; LD: lipid droplet; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MG: monoglyceride; MOI: multiplicity of infection; mRFP: monomeric red fluorescent protein; NR1D2: nuclear receptor subfamily 1 group D member 2; p.i., post-infection; PLIN2/ADRP: perilipin 2; PNPLA2/ATGL: patatin like phospholipase domain containing 2; Rapa: rapamycin; SQSTM1/p62: sequestosome 1; shRNA: short hairpin RNA; TEM: transmission electron microscopy; TG: triglyceride.
Collapse
Affiliation(s)
- Mengling Tang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China.,Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Zhiqiang Hu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chenglong Rao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiangao Chen
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China.,Department of General Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Siqi Yuan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China.,Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Jiangang Zhang
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chan Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingmin Yan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yupei Xia
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Meijuan Zhang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Juanjuan Yue
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yang Xiang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Xuhu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
21
|
Redundant and Cooperative Roles for Yersinia pestis Yop Effectors in the Inhibition of Human Neutrophil Exocytic Responses Revealed by Gain-of-Function Approach. Infect Immun 2020; 88:IAI.00909-19. [PMID: 31871100 DOI: 10.1128/iai.00909-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Yersinia pestis causes a rapid, lethal disease referred to as plague. Y. pestis actively inhibits the innate immune system to generate a noninflammatory environment during early stages of infection to promote colonization. The ability of Y. pestis to create this early noninflammatory environment is in part due to the action of seven Yop effector proteins that are directly injected into host cells via a type 3 secretion system (T3SS). While each Yop effector interacts with specific host proteins to inhibit their function, several Yop effectors either target the same host protein or inhibit converging signaling pathways, leading to functional redundancy. Previous work established that Y. pestis uses the T3SS to inhibit neutrophil respiratory burst, phagocytosis, and release of inflammatory cytokines. Here, we show that Y. pestis also inhibits release of granules in a T3SS-dependent manner. Moreover, using a gain-of-function approach, we discovered previously hidden contributions of YpkA and YopJ to inhibition and that cooperative actions by multiple Yop effectors are required to effectively inhibit degranulation. Independent from degranulation, we also show that multiple Yop effectors can inhibit synthesis of leukotriene B4 (LTB4), a potent lipid mediator released by neutrophils early during infection to promote inflammation. Together, inhibition of these two arms of the neutrophil response likely contributes to the noninflammatory environment needed for Y. pestis colonization and proliferation.
Collapse
|
22
|
Immunomodulatory activity of hyaluronidase is associated with metabolic adaptations during acute inflammation. Inflamm Res 2019; 69:105-113. [DOI: 10.1007/s00011-019-01297-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/10/2019] [Accepted: 10/31/2019] [Indexed: 12/31/2022] Open
|
23
|
Pereira-Dutra FS, Teixeira L, de Souza Costa MF, Bozza PT. Fat, fight, and beyond: The multiple roles of lipid droplets in infections and inflammation. J Leukoc Biol 2019; 106:563-580. [PMID: 31121077 DOI: 10.1002/jlb.4mr0119-035r] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/16/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
Increased accumulation of cytoplasmic lipid droplets (LDs) in host nonadipose cells is commonly observed in response to numerous infectious diseases, including bacterial, parasite, and fungal infections. LDs are lipid-enriched, dynamic organelles composed of a core of neutral lipids surrounded by a monolayer of phospholipids associated with a diverse array of proteins that are cell and stimulus regulated. Far beyond being simply a deposit of neutral lipids, LDs have come to be seen as an essential platform for various cellular processes, including metabolic regulation, cell signaling, and the immune response. LD participation in the immune response occurs as sites for compartmentalization of several immunometabolic signaling pathways, production of inflammatory lipid mediators, and regulation of antigen presentation. Infection-driven LD biogenesis is a complexly regulated process that involves innate immune receptors, transcriptional and posttranscriptional regulation, increased lipid uptake, and new lipid synthesis. Accumulating evidence demonstrates that intracellular pathogens are able to exploit LDs as an energy source, a replication site, and/or a mechanism of immune response evasion. Nevertheless, LDs can also act in favor of the host as part of the immune and inflammatory response to pathogens. Here, we review recent findings that explored the new roles of LDs in the context of host-pathogen interactions.
Collapse
Affiliation(s)
- Filipe S Pereira-Dutra
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Livia Teixeira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Maoduo Z, Hao Y, Wei W, Feng W, Dagan M. Effects of LPS on the accumulation of lipid droplets, proliferation, and steroidogenesis in goat luteinized granulosa cells. J Biochem Mol Toxicol 2019; 33:e22329. [PMID: 30934154 DOI: 10.1002/jbt.22329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/25/2019] [Accepted: 03/15/2019] [Indexed: 12/26/2022]
Abstract
Lipopolysaccharide (LPS) can cause ovarian dysfunction and infertility in mammals. The purpose of this study was to investigate the effects of LPS on the accumulation of lipid droplets (LDs), proliferation, and steroidogenesis in goat luteinized granulosa cells (LGCs). GCs isolated from the ovarian follicles were spontaneously luteinized under media with fetal bovine serum, resulting in increased progesterone and shifted shape from spherical to star with multiple prolongations. Then, LGCs were treated with LPS (0-10 μg/mL) for 0-48 hours. Oil Red O staining was performed to observe LDs accumulation and commercial kit was applied to detect intracellular triglyceride (TG) content. The cell proliferation were detected by cell counting kit-8. Expressions of cell-cycle-related genes were determined by real-time polymerase chain reaction. Estradiol (E 2 ) and progesterone (P 4 ) from cell supernatants were determined by enzyme-linked immunosorbent assay, and expressions of STAR, P450scc, 3β-hydroxysteroid dehydrogenase (3β-HSD) and CYP19A1 were detected by Western blot. Results showed that LPS treatment significantly increased LDs accumulation after 24 hours, and 5 μg/mL LPS increased TG content ( P < 0.05). LPS treatment for 24 hours stimulated the LGCs activities ( P<0.05), which was confirmed by the increases in the expressions of proliferating cell nuclear antigen (PCNA), cyclinB1 and cyclinD1, while 48 hours treatment had no effect. LPS treatment suppressed E 2 and P 4 output of LGCs ( P < 0.05). Western blot results showed that 10 μg/mL LPS decreased the protein expression of 3β-HSD in LGCs ( P < 0.05). In conclusion, LPS increased LDs accumulation and cell proliferation, and LPS-mediated P 4 reduction could be attributed to the decreased 3β-HSD protein expression, which provide new information for the regulation of ovarian function in goats.
Collapse
Affiliation(s)
- Zhang Maoduo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - Yu Hao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - Wang Wei
- Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - Wang Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - Mao Dagan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
25
|
Töpfer N, Müller MM, Dahms M, Ramoji A, Popp J, Slevogt H, Neugebauer U. Raman spectroscopy reveals LPS-induced changes of biomolecular composition in monocytic THP-1 cells in a label-free manner. Integr Biol (Camb) 2019; 11:87-98. [PMID: 31083720 DOI: 10.1093/intbio/zyz009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/01/2019] [Accepted: 03/20/2019] [Indexed: 12/23/2022]
Abstract
The human innate immune system is able to recognize pathogen-associated molecular patterns like lipopolysaccharides (LPS) leading to the activation of signal cascades and the release of different cytokines. Activation of the immune cells can be assessed in different ways which are either indirect (ELISA of cytokine release), require staining protocols (flow cytometry) or lysis of the cells (mRNA analysis). Here, Raman spectroscopy as a non-destructive spectroscopic method is presented to enable direct and label-free monitoring of changes in cellular metabolism, biomolecular composition as well as morphology. Exemplarily, the potential of Raman spectroscopy is presented for the characterization of LPS-stimulation of monocytic THP-1 cells over a time course of 16 h. The cell culture stimulation model is characterized using gene transcription and expression of the two cytokines TNFα and IL-1β. After 1 h, 3 h, 8 h and 16 h specific Raman spectroscopic fingerprints are generated which encode cell activation pattern after TLR4 stimulation. Most prevalent changes in the spectra occur after 8 h, but slight differences are already detectable after 1 h. Spatially highly resolved Raman scans are used to generate false-color Raman images which provide spatial information of the biochemical state of the cells and changes over time. One of the most significant observed differences is an increase in Raman signal from DNA/RNA content in LPS-stimulated cells when compared to unstimulated cells. The systematic assignment of Raman spectroscopic profiles of LPS-activated cells to cellular activation assessed by cytokine gene transcription and expression opens new ways for label-free and direct immunological studies of specific pathogen recognizing receptors and their downstream signaling pathways.
Collapse
Affiliation(s)
- Natalie Töpfer
- Leibniz Institute of Photonic Technology, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Mario M Müller
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Host Septomics, Jena University Hospital, Jena, Germany
| | - Marcel Dahms
- Leibniz Institute of Photonic Technology, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Institute for Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, Jena, Germany
- InfectoGnostics Research Campus Jena, Reg. Assoc., Jena, Germany
| | - Anuradha Ramoji
- Leibniz Institute of Photonic Technology, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Institute for Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, Jena, Germany
- InfectoGnostics Research Campus Jena, Reg. Assoc., Jena, Germany
| | - Hortense Slevogt
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Host Septomics, Jena University Hospital, Jena, Germany
| | - Ute Neugebauer
- Leibniz Institute of Photonic Technology, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Institute for Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, Jena, Germany
- InfectoGnostics Research Campus Jena, Reg. Assoc., Jena, Germany
| |
Collapse
|
26
|
Phelan JJ, Basdeo SA, Tazoll SC, McGivern S, Saborido JR, Keane J. Modulating Iron for Metabolic Support of TB Host Defense. Front Immunol 2018; 9:2296. [PMID: 30374347 PMCID: PMC6196273 DOI: 10.3389/fimmu.2018.02296] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/17/2018] [Indexed: 02/05/2023] Open
Abstract
Tuberculosis (TB) is the world's biggest infectious disease killer. The increasing prevalence of multidrug-resistant and extensively drug-resistant TB demonstrates that current treatments are inadequate and there is an urgent need for novel therapies. Research is now focused on the development of host-directed therapies (HDTs) which can be used in combination with existing antimicrobials, with a special focus on promoting host defense. Immunometabolic reprogramming is integral to TB host defense, therefore, understanding and supporting the immunometabolic pathways that are altered after infection will be important for the development of new HDTs. Moreover, TB pathophysiology is interconnected with iron metabolism. Iron is essential for the survival of Mycobacterium tuberculosis (Mtb), the bacteria that causes TB disease. Mtb struggles to replicate and persist in low iron environments. Iron chelation has therefore been suggested as a HDT. In addition to its direct effects on iron availability, iron chelators modulate immunometabolism through the stabilization of HIF1α. This review examines immunometabolism in the context of Mtb and its links to iron metabolism. We suggest that iron chelation, and subsequent stabilization of HIF1α, will have multifaceted effects on immunometabolic function and holds potential to be utilized as a HDT to boost the host immune response to Mtb infection.
Collapse
Affiliation(s)
- James J Phelan
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Sharee A Basdeo
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Simone C Tazoll
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Sadhbh McGivern
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Judit R Saborido
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| |
Collapse
|
27
|
Mitochondrial-Targeted Antioxidant MitoQ Prevents E. coli Lipopolysaccharide-Induced Accumulation of Triacylglycerol and Lipid Droplets Biogenesis in Epithelial Cells. J Lipids 2018; 2018:5745790. [PMID: 30245885 PMCID: PMC6139225 DOI: 10.1155/2018/5745790] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/02/2018] [Accepted: 07/25/2018] [Indexed: 01/26/2023] Open
Abstract
The effect of bacterial lipopolysaccharide (LPS) on eukaryotic cell could be accompanied by a significant metabolic shift that includes accumulation of triacylglycerol (TAG) in lipid droplets (LD), ubiquitous organelles associated with fatty acid storage, energy regulation and demonstrated tight spatial and functional connections with mitochondria. The impairment of mitochondrial activity under pathological stimuli has been shown to provoke TAG storage and LD biogenesis. However the potential mechanisms that link mitochondrial disturbances and TAG accumulation are not completely understood. We hypothesize that mitochondrial ROS (mROS) may play a role of a trigger leading to subsequent accumulation of intracellular TAG and LD in response to a bacterial stimulus. Using isolated epithelial cells from the frog urinary bladder, we showed that LPS decreased fatty acids oxidation, enhanced TAG deposition, and promoted LD formation. LPS treatment did not affect the mitochondrial membrane potential but increased cellular ROS production and led to impairment of mitochondrial function as revealed by decreased ATP production and a reduced maximal oxygen consumption rate (OCR) and OCR directed at ATP turnover. The mitochondrial-targeted antioxidant MitoQ at a dose of 25 nM did not prevent LPS-induced alterations in cellular respiration, but, in contrast to nonmitochondrial antioxidant α-tocopherol, reduced the effect of LPS on the generation of ROS, restored the LPS-induced decline of fatty acids oxidation, and prevented accumulation of TAG and LD biogenesis. The data obtained indicate the key signaling role of mROS in the lipid metabolic shift that occurs under the impact of a bacterial pathogen in epithelial cells.
Collapse
|
28
|
Vallochi AL, Teixeira L, Oliveira KDS, Maya-Monteiro CM, Bozza PT. Lipid Droplet, a Key Player in Host-Parasite Interactions. Front Immunol 2018; 9:1022. [PMID: 29875768 PMCID: PMC5974170 DOI: 10.3389/fimmu.2018.01022] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/24/2018] [Indexed: 12/18/2022] Open
Abstract
Lipid droplets (lipid bodies, LDs) are dynamic organelles that have important roles in regulating lipid metabolism, energy homeostasis, cell signaling, membrane trafficking, and inflammation. LD biogenesis, composition, and functions are highly regulated and may vary according to the stimuli, cell type, activation state, and inflammatory environment. Increased cytoplasmic LDs are frequently observed in leukocytes and other cells in a number of infectious diseases. Accumulating evidence reveals LDs participation in fundamental mechanisms of host-pathogen interactions, including cell signaling and immunity. LDs are sources of eicosanoid production, and may participate in different aspects of innate signaling and antigen presentation. In addition, intracellular pathogens evolved mechanisms to subvert host metabolism and may use host LDs, as ways of immune evasion and nutrients source. Here, we review mechanisms of LDs biogenesis and their contributions to the infection progress, and discuss the latest discoveries on mechanisms and pathways involving LDs roles as regulators of the immune response to protozoan infection.
Collapse
Affiliation(s)
- Adriana Lima Vallochi
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | | | | | - Patricia T. Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
29
|
de Almeida PE, Toledo DAM, Rodrigues GSC, D'Avila H. Lipid Bodies as Sites of Prostaglandin E2 Synthesis During Chagas Disease: Impact in the Parasite Escape Mechanism. Front Microbiol 2018; 9:499. [PMID: 29616011 PMCID: PMC5869919 DOI: 10.3389/fmicb.2018.00499] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/05/2018] [Indexed: 12/22/2022] Open
Abstract
During Chagas disease, the Trypanosoma cruzi can induce some changes in the host cells in order to escape or manipulate the host immune response. The modulation of the lipid metabolism in the host phagocytes or in the parasite itself is one feature that has been observed. The goal of this mini review is to discuss the mechanisms that regulate intracellular lipid body (LB) biogenesis in the course of this parasite infection and their meaning to the pathophysiology of the disease. The interaction host–parasite induces LB (or lipid droplet) formation in a Toll-like receptor 2-dependent mechanism in macrophages and is enhanced by apoptotic cell uptake. Simultaneously, there is a lipid accumulation in the parasite due to the incorporation of host fatty acids. The increase in the LB accumulation during infection is correlated with an increase in the synthesis of PGE2 within the host cells and the parasite LBs. Moreover, the treatment with fatty acid synthase inhibitor C75 or non-steroidal anti-inflammatory drugs such as NS-398 and aspirin inhibited the LB biogenesis and also induced the down modulation of the eicosanoid production and the parasite replication. These findings show that LBs are organelles up modulated during the course of infection. Furthermore, the biogenesis of the LB is involved in the lipid mediator generation by both the macrophages and the parasite triggering escape mechanisms.
Collapse
Affiliation(s)
- Patrícia E de Almeida
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Gabriel S C Rodrigues
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Heloisa D'Avila
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
30
|
Teng O, Ang CKE, Guan XL. Macrophage-Bacteria Interactions-A Lipid-Centric Relationship. Front Immunol 2017; 8:1836. [PMID: 29326713 PMCID: PMC5742358 DOI: 10.3389/fimmu.2017.01836] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/05/2017] [Indexed: 11/13/2022] Open
Abstract
Macrophages are professional phagocytes at the front line of immune defenses against foreign bodies and microbial pathogens. Various bacteria, which are responsible for deadly diseases including tuberculosis and salmonellosis, are capable of hijacking this important immune cell type and thrive intracellularly, either in the cytoplasm or in specialized vacuoles. Tight regulation of cellular metabolism is critical in shaping the macrophage polarization states and immune functions. Lipids, besides being the bulk component of biological membranes, serve as energy sources as well as signaling molecules during infection and inflammation. With the advent of systems-scale analyses of genes, transcripts, proteins, and metabolites, in combination with classical biology, it is increasingly evident that macrophages undergo extensive lipid remodeling during activation and infection. Each bacterium species has evolved its own tactics to manipulate host metabolism toward its own advantage. Furthermore, modulation of host lipid metabolism affects disease susceptibility and outcome of infections, highlighting the critical roles of lipids in infectious diseases. Here, we will review the emerging roles of lipids in the complex host-pathogen relationship and discuss recent methodologies employed to probe these versatile metabolites during the infection process. An improved understanding of the lipid-centric nature of infections can lead to the identification of the Achilles' heel of the pathogens and host-directed targets for therapeutic interventions. Currently, lipid-moderating drugs are clinically available for a range of non-communicable diseases, which we anticipate can potentially be tapped into for various infections.
Collapse
Affiliation(s)
- Ooiean Teng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Candice Ke En Ang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Xue Li Guan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
31
|
Abstract
Bioactive lipids regulate most physiological processes, from digestion to blood flow and from hemostasis to labor. Lipid mediators are also involved in multiple pathologies including cancer, autoimmunity or asthma. The pathological roles of lipid mediators are based on their intricate involvement in the immune system, which comprises source and target cells of these mediators. Based on their biosynthetic origin, bioactive lipids can be grouped into different classes [e.g. sphingolipids, formed from sphingosine or eicosanoids, formed from arachidonic acid (AA)]. Owing to the complexity of different mediator classes and the prominent immunological roles of eicosanoids, this review will focus solely on the immune-regulation of eicosanoids. Eicosanoids do not only control key immune responses (e.g. chemotaxis, antigen presentation, phagocytosis), but they are also subject to reciprocal control by the immune system. Particularly, key immunoregulatory cytokines such as IL-4 and IFN-γ shape the cellular eicosanoid profile, thus providing efficient feedback regulation between cytokine and eicosanoid networks. For the purpose of this review, I will first provide a short overview of the most important immunological functions of eicosanoids with a focus on prostaglandins (PGs) and leukotrienes (LTs). Second, I will summarize the current knowledge on immunological factors that regulate eicosanoid production during infection and inflammation.
Collapse
|
32
|
Jin SH, An SK, Lee SB. The formation of lipid droplets favors intracellular Mycobacterium leprae survival in SW-10, non-myelinating Schwann cells. PLoS Negl Trop Dis 2017. [PMID: 28636650 PMCID: PMC5495515 DOI: 10.1371/journal.pntd.0005687] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Leprosy is a chronic infectious disease that is caused by the obligate intracellular pathogen Mycobacterium leprae (M.leprae), which is the leading cause of all non-traumatic peripheral neuropathies worldwide. Although both myelinating and non-myelinating Schwann cells are infected by M.leprae in patients with lepromatous leprosy, M.leprae preferentially invades the non-myelinating Schwann cells. However, the effect of M.leprae infection on non-myelinating Schwann cells has not been elucidated. Lipid droplets (LDs) are found in M.leprae-infected Schwann cells in the nerve biopsies of lepromatous leprosy patients. M.leprae-induced LD formation favors intracellular M.leprae survival in primary Schwann cells and in a myelinating Schwann cell line referred to as ST88-14. In the current study, we initially characterized SW-10 cells and investigated the effects of LDs on M.leprae-infected SW-10 cells, which are non-myelinating Schwann cells. SW-10 cells express S100, a marker for cells from the neural crest, and NGFR p75, a marker for immature or non-myelinating Schwann cells. SW-10 cells, however, do not express myelin basic protein (MBP), a marker for myelinating Schwann cells, and myelin protein zero (MPZ), a marker for precursor, immature, or myelinating Schwann cells, all of which suggests that SW-10 cells are non-myelinating Schwann cells. In addition, SW-10 cells have phagocytic activity and can be infected with M. leprae. Infection with M. leprae induces the formation of LDs. Furthermore, inhibiting the formation of M. leprae-induced LD enhances the maturation of phagosomes containing live M.leprae and decreases the ATP content in the M. leprae found in SW-10 cells. These facts suggest that LD formation by M. leprae favors intracellular M. leprae survival in SW-10 cells, which leads to the logical conclusion that M.leprae-infected SW-10 cells can be a new model for investigating the interaction of M.leprae with non-myelinating Schwann cells. Leprosy is a chronic infectious disease that is caused by the obligate intracellular pathogen Mycobacterium leprae (M.leprae). Leprosy is the leading cause of all non-traumatic peripheral neuropathies worldwide. Both myelinating and non-myelinating Schwann cells are infected by M.leprae in lepromatous leprosy, but the non-myelinating Schwann cells show greater susceptibility to M.leprae invasion. However, the effect of M.leprae infection on non-myelinating Schwann cells has not been elucidated. Our results show that SW-10 cells are non-myelinating Schwann cells. Infection with M. leprae induces lipid droplet (LD) formation. Furthermore, inhibition of M. leprae-induced LD formation enhances the maturation of phagosomes containing live M.leprae and decreases the ATP content of M. leprae in SW-10 cells, suggesting that LD formation by M. leprae favors M. leprae survival in SW-10 cells. Based on these findings, it should be clear that M.leprae-infected SW-10 cells can serve as a new model for investigating the interaction of M.leprae with non-myelinating Schwann cells.
Collapse
Affiliation(s)
- Song-Hyo Jin
- Institute of Hansen’s Disease, Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, Republic of Korea
- Research Institute for Molecular-targeted Drugs, Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Sung-Kwan An
- Research Institute for Molecular-targeted Drugs, Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Seong-Beom Lee
- Institute of Hansen’s Disease, Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
33
|
Roingeard P, Melo RCN. Lipid droplet hijacking by intracellular pathogens. Cell Microbiol 2016; 19. [PMID: 27794207 DOI: 10.1111/cmi.12688] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 10/24/2016] [Accepted: 10/26/2016] [Indexed: 12/15/2022]
Abstract
Lipid droplets were long considered to be simple storage structures, but they have recently been shown to be dynamic organelles involved in diverse biological processes, including emerging roles in innate immunity. Various intracellular pathogens, including viruses, bacteria, and parasites, specifically target host lipid droplets during their life cycle. Viruses such as hepatitis C, dengue, and rotaviruses use lipid droplets as platforms for assembly. Bacteria, such as mycobacteria and Chlamydia, and parasites, such as trypanosomes, use host lipid droplets for nutritional purposes. The possible use of lipid droplets by intracellular pathogens, as part of an anti-immunity strategy, is an intriguing question meriting further investigation in the near future.
Collapse
Affiliation(s)
- Philippe Roingeard
- INSERM U966 and IBiSA Electron Microscopy Facility, François Rabelais University and CHRU de Tours, Tours, France
| | - Rossana C N Melo
- Laboratory of Cell Biology, Institute of Biological Sciences, Federal University of Juiz de Fora-UFJF, Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
34
|
Xue H, Yang RY, Tai G, Liu FT. Galectin-12 inhibits granulocytic differentiation of human NB4 promyelocytic leukemia cells while promoting lipogenesis. J Leukoc Biol 2016; 100:657-664. [PMID: 27256573 DOI: 10.1189/jlb.1hi0316-134r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/29/2016] [Indexed: 12/19/2022] Open
Abstract
As a member of the galectin family of animal lectins, galectin-12 is preferentially expressed in adipocytes and leukocytes. In adipocytes, galectin-12 is associated with lipid droplets and regulates lipid metabolism and energy balance, whereas its role in leukocytes is not clear. Analysis of galectin-12 expression in a public data set of acute myeloid leukemia (AML) samples revealed that it is selectively overexpressed in the M3 subtype, which is also known as acute promyelocytic leukemia (APL). To investigate the role of galectin-12 in APL cells, we manipulated its expression in the APL cell line, NB4, and measured resultant effects on all-trans-retinoic acid (ATRA)-induced granulocytic differentiation. With a doxycycline-inducible gene knockdown system, we found that suppression of galectin-12 promoted ATRA-induced neutrophil differentiation but inhibited lipid droplet formation. Our results indicate that overexpression of galectin-12 contributes to a differentiation block in APL cells, and suppression of galectin-12 facilitates granulocytic differentiation. Furthermore, these data suggest that lipogenesis and other aspects of myeloid differentiation can be differentially regulated. Taken together, these findings suggest that galectin-12 may be a target for treatment of the ATRA-resistant subset of APL.
Collapse
Affiliation(s)
- Huiting Xue
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, USA; School of Life Sciences, Northeast Normal University, Changchun, China; and
| | - Ri-Yao Yang
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, USA
| | - Guihua Tai
- School of Life Sciences, Northeast Normal University, Changchun, China; and
| | - Fu-Tong Liu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, USA; Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, Taiwan
| |
Collapse
|
35
|
Barletta ABF, Alves LR, Silva MCLN, Sim S, Dimopoulos G, Liechocki S, Maya-Monteiro CM, Sorgine MHF. Emerging role of lipid droplets in Aedes aegypti immune response against bacteria and Dengue virus. Sci Rep 2016; 6:19928. [PMID: 26887863 PMCID: PMC4757862 DOI: 10.1038/srep19928] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/21/2015] [Indexed: 12/19/2022] Open
Abstract
In mammals, lipid droplets (LDs) are ubiquitous organelles that modulate immune and inflammatory responses through the production of lipid mediators. In insects, it is unknown whether LDs play any role during the development of immune responses. We show that Aedes aegypti Aag2 cells – an immune responsive cell lineage – accumulates LDs when challenged with Enterobacter cloacae, Sindbis, and Dengue viruses. Microarray analysis of Aag2 challenged with E.cloacae or infected with Dengue virus revealed high transcripts levels of genes associated with lipid storage and LDs biogenesis, correlating with the increased LDs numbers in those conditions. Similarly, in mosquitoes, LDs accumulate in midgut cells in response to Serratia marcescens and Sindbis virus or when the native microbiota proliferates, following a blood meal. Also, constitutive activation of Toll and IMD pathways by knocking-down their respective negative modulators (Cactus and Caspar) increases LDs numbers in the midgut. Our results show for the first time an infection-induced LDs accumulation in response to both bacterial and viral infections in Ae. Aegypti, and we propose a role for LDs in mosquito immunity. These findings open new venues for further studies in insect immune responses associated with lipid metabolism.
Collapse
Affiliation(s)
- Ana Beatriz Ferreira Barletta
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo De Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Brasil
| | - Liliane Rosa Alves
- Instituto Nacional de Câncer, Hospital do Câncer II, Rio de Janeiro, RJ, Brasil
| | - Maria Clara L Nascimento Silva
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo De Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Shuzhen Sim
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sally Liechocki
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brasil
| | - Clarissa M Maya-Monteiro
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brasil
| | - Marcos H Ferreira Sorgine
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo De Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Brasil
| |
Collapse
|
36
|
Li X, Xue W, Liu Y, Li W, Fan D, Zhu C, Wang Y. HLC/pullulan and pullulan hydrogels: their microstructure, engineering process and biocompatibility. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 58:1046-57. [DOI: 10.1016/j.msec.2015.09.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/07/2015] [Indexed: 01/21/2023]
|
37
|
Greineisen WE, Maaetoft-Udsen K, Speck M, Balajadia J, Shimoda LMN, Sung C, Turner H. Chronic Insulin Exposure Induces ER Stress and Lipid Body Accumulation in Mast Cells at the Expense of Their Secretory Degranulation Response. PLoS One 2015; 10:e0130198. [PMID: 26263026 PMCID: PMC4532411 DOI: 10.1371/journal.pone.0130198] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 05/17/2015] [Indexed: 12/11/2022] Open
Abstract
Lipid bodies (LB) are reservoirs of precursors to inflammatory lipid mediators in immunocytes, including mast cells. LB numbers are dynamic, increasing dramatically under conditions of immunological challenge. We have previously shown in vitro that insulin-influenced lipogenic pathways induce LB biogenesis in mast cells, with their numbers attaining steatosis-like levels. Here, we demonstrate that in vivo hyperinsulinemia resulting from high fat diet is associated with LB accumulation in murine mast cells and basophils. We characterize the lipidome of purified insulin-induced LB, and the shifts in the whole cell lipid landscape in LB that are associated with their accumulation, in both model (RBL2H3) and primary mast cells. Lipidomic analysis suggests a gain of function associated with LB accumulation, in terms of elevated levels of eicosanoid precursors that translate to enhanced antigen-induced LTC4 release. Loss-of-function in terms of a suppressed degranulation response was also associated with LB accumulation, as were ER reprogramming and ER stress, analogous to observations in the obese hepatocyte and adipocyte. Taken together, these data suggest that chronic insulin elevation drives mast cell LB enrichment in vitro and in vivo, with associated effects on the cellular lipidome, ER status and pro-inflammatory responses.
Collapse
Affiliation(s)
- William E. Greineisen
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawaii, United States of America
| | - Kristina Maaetoft-Udsen
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawaii, United States of America
| | - Mark Speck
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawaii, United States of America
| | - Januaria Balajadia
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawaii, United States of America
| | - Lori M. N. Shimoda
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawaii, United States of America
| | - Carl Sung
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawaii, United States of America
| | - Helen Turner
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawaii, United States of America
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
38
|
Arisqueta L, Navarro-Imaz H, Rueda Y, Fresnedo O. Cholesterol mobilization from hepatic lipid droplets during endotoxemia is altered in obese ob/ob mice. J Biochem 2015; 158:321-9. [DOI: 10.1093/jb/mvv047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/31/2015] [Indexed: 11/12/2022] Open
|
39
|
Blockade of the JNK signalling as a rational therapeutic approach to modulate the early and late steps of the inflammatory cascade in polymicrobial sepsis. Mediators Inflamm 2015; 2015:591572. [PMID: 25873765 PMCID: PMC4385695 DOI: 10.1155/2015/591572] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/26/2014] [Indexed: 01/12/2023] Open
Abstract
Cecal ligation and puncture (CLP) is an experimental polymicrobial sepsis induced systemic inflammation that leads to acute organ failure. Aim of our study was to evaluate the effects of SP600125, a specific c-Jun NH2-terminal kinase (JNK) inhibitor, to modulate the early and late steps of the inflammatory cascade in a murine model of CLP-induced sepsis. CB57BL/6J mice were subjected to CLP or sham operation. Animals were randomized to receive either SP600125 (15 mg/kg) or its vehicle intraperitoneally 1 hour after surgery and repeat treatment every 24 hours. To evaluate survival, a group of animals was monitored every 24 hours for 120 hours. Two other animals were sacrificed 4 or 18 hours after surgical procedures; lung and liver samples were collected for biomolecular and histopathologic analysis. The expression of p-JNK, p-ERK, TNF-α, HMGB-1, NF-κB, Ras, Rho, Caspase 3, Bcl-2, and Bax was evaluated in lung and liver samples; SP600125 improved survival, reduced CLP induced activation of JNK, NF-κB, TNF-α, and HMGB-1, inhibited proapoptotic pathway, preserved Bcl-2 expression, and reduced histologic damage in both lung and liver of septic mice. SP600125 protects against CLP induced sepsis by blocking JNK signalling; therefore, it can be considered a therapeutic approach in human sepsis.
Collapse
|
40
|
Graner MW, Lillehei KO, Katsanis E. Endoplasmic reticulum chaperones and their roles in the immunogenicity of cancer vaccines. Front Oncol 2015; 4:379. [PMID: 25610811 PMCID: PMC4285071 DOI: 10.3389/fonc.2014.00379] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/17/2014] [Indexed: 11/25/2022] Open
Abstract
The endoplasmic reticulum (ER) is a major site of passage for proteins en route to other organelles, to the cell surface, and to the extracellular space. It is also the transport route for peptides generated in the cytosol by the proteasome into the ER for loading onto major histocompatibility complex class I (MHC I) molecules for eventual antigen presentation at the cell surface. Chaperones within the ER are critical for many of these processes; however, outside the ER certain of those chaperones may play important and direct roles in immune responses. In some cases, particular ER chaperones have been utilized as vaccines against tumors or infectious disease pathogens when purified from tumor tissue or recombinantly generated and loaded with antigen. In other cases, the cell surface location of ER chaperones has implications for immune responses as well as possible tumor resistance. We have produced heat-shock protein/chaperone protein-based cancer vaccines called “chaperone-rich cell lysate” (CRCL) that are conglomerates of chaperones enriched from solid tumors by an isoelectric focusing technique. These preparations have been effective against numerous murine tumors, as well as in a canine with an advanced lung carcinoma treated with autologous CRCL. We also published extensive proteomic analyses of CRCL prepared from human surgically resected tumor samples. Of note, these preparations contained at least 10 ER chaperones and a number of other residents, along with many other chaperones/heat-shock proteins. Gene ontology and network analyses utilizing these proteins essentially recapitulate the antigen presentation pathways and interconnections. In conjunction with our current knowledge of cell surface/extracellular ER chaperones, these data collectively suggest that a systems-level view may provide insight into the potent immune stimulatory activities of CRCL with an emphasis on the roles of ER components in those processes.
Collapse
Affiliation(s)
- Michael W Graner
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado School of Medicine , Aurora, CO , USA
| | - Kevin O Lillehei
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado School of Medicine , Aurora, CO , USA
| | - Emmanuel Katsanis
- Department of Pediatrics, The University of Arizona , Tucson, AZ , USA
| |
Collapse
|
41
|
Maaetoft-Udsen K, Greineisen WE, Aldan JT, Magaoay H, Ligohr C, Shimoda LMN, Sung C, Turner H. Comparative analysis of lipotoxicity induced by endocrine, pharmacological, and innate immune stimuli in rat basophilic leukemia cells. J Immunotoxicol 2014; 12:385-94. [PMID: 25539471 DOI: 10.3109/1547691x.2014.990655] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cellular lipotoxicity manifests as the steatotic accumulation of lipid droplets or lipid bodies, and/or induction of phospholipidosis. Lipotoxicity can be induced by hyperinsulinemia/nutrient overload, cationic amphiphilic drugs (CAD), and innate immunological stimuli, all of which are stimuli relevant to mast cell physiology. Hyper-accumulation of mast cell lipid bodies in response to hyperinsulinemia has been documented, but lipotoxicity in response to CAD or innate immunologic stimuli has not been analysed comparatively. Moreover, gaps in our understanding of this steatosis remain, specifically as to whether hyperinsulinemia-driven steatosis in these cells attains lipotoxic levels or is accompanied by phospholipidosis. To compare endocrine, pharmacological, and innate immunological stimuli for their ability to induce steatosis and phospholipidosis in a rat basophilic leukemia mast cell model (RBL2H3), differential fluorescence microscopy staining and quantitation of phospholipidosis and steatosis in the RBL2H3 cell line was examined. The three classes of stimuli differentially induced phospholipidosis and steatosis. PPARγ up-regulation was not uniformly associated with the expansion of the lipid body population. Fluorescence imaging of lipid-enriched structures generated in response to lipotoxic cationic amphiphilic drugs, chronic insulin exposure, and TLR2/4 ligands revealed differential staining patterns when visualized using lipophilic dyes. It is concluded that lipotoxicity-inducing pathways in this model mast cell system are diverse, and include steatotic responses to an endocrine stimulus, as well as phospholipidosis responses to cationic lipophilic drugs not previously described in this cell type.
Collapse
|
42
|
Zoccal KF, Paula-Silva FWG, Bitencourt CDS, Sorgi CA, Bordon KDCF, Arantes EC, Faccioli LH. PPAR-γ activation by Tityus serrulatus venom regulates lipid body formation and lipid mediator production. Toxicon 2014; 93:90-7. [PMID: 25450800 DOI: 10.1016/j.toxicon.2014.11.226] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/31/2014] [Accepted: 11/13/2014] [Indexed: 02/06/2023]
Abstract
Tityus serrulatus venom (TsV) consists of numerous peptides with different physiological and pharmacological activities. Studies have shown that scorpion venom increases pro-inflammatory cytokine production, contributing to immunological imbalance, multiple organ dysfunction, and patient death. We have previously demonstrated that TsV is a venom-associated molecular pattern (VAMP) recognized by TLRs inducing intense inflammatory reaction through the production of pro-inflammatory cytokines and arachidonic acid-derived lipid mediators prostaglandin (PG)E2 and leukotriene (LT)B4. Lipid bodies (LBs) are potential sites for eicosanoid production by inflammatory cells. Moreover, recent studies have shown that the peroxisome proliferator-activated receptor gamma (PPAR-γ) is implicated in LB formation and acts as an important modulator of lipid metabolism during inflammation. In this study, we used murine macrophages to evaluate whether the LB formation induced by TsV after TLR recognition correlates with lipid mediator generation by macrophages and if it occurs through PPAR-γ activation. We demonstrate that TsV acts through TLR2 and TLR4 stimulation and PPAR-γ activation to induce LB formation and generation of PGE2 and LTB4. Our data also show that PPAR-γ negatively regulates the pro-inflammatory NF-κB transcription factor. Based on these results, we suggest that during envenomation, LBs constitute functional organelles for lipid mediator production through signaling pathways that depend on cell surface and nuclear receptors. These findings point to the inflammatory mechanisms that might also be triggered during human envenomation by TsV.
Collapse
Affiliation(s)
- Karina Furlani Zoccal
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Brazil.
| | - Francisco Wanderley Garcia Paula-Silva
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Brazil.
| | - Claudia da Silva Bitencourt
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Brazil.
| | - Carlos Artério Sorgi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Brazil.
| | | | - Eliane Candiani Arantes
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Brazil.
| | - Lúcia Helena Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Brazil.
| |
Collapse
|
43
|
Moon TC, Befus AD, Kulka M. Mast cell mediators: their differential release and the secretory pathways involved. Front Immunol 2014; 5:569. [PMID: 25452755 PMCID: PMC4231949 DOI: 10.3389/fimmu.2014.00569] [Citation(s) in RCA: 291] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 10/23/2014] [Indexed: 12/14/2022] Open
Abstract
Mast cells (MC) are widely distributed throughout the body and are common at mucosal surfaces, a major host-environment interface. MC are functionally and phenotypically heterogeneous depending on the microenvironment in which they mature. Although MC have been classically viewed as effector cells of IgE-mediated allergic diseases, they are also recognized as important in host defense, innate and acquired immunity, homeostatic responses, and immunoregulation. MC activation can induce release of pre-formed mediators such as histamine from their granules, as well as release of de novo synthesized lipid mediators, cytokines, and chemokines that play diverse roles, not only in allergic reactions but also in numerous physiological and pathophysiological responses. Indeed, MC release their mediators in a discriminating and chronological manner, depending upon the stimuli involved and their signaling cascades (e.g., IgE-mediated or Toll-like receptor-mediated). However, the precise mechanisms underlying differential mediator release in response to these stimuli are poorly known. This review summarizes our knowledge of MC mediators and will focus on what is known about the discriminatory release of these mediators dependent upon diverse stimuli, MC phenotypes, and species of origin, as well as on the intracellular synthesis, storage, and secretory processes involved.
Collapse
Affiliation(s)
- Tae Chul Moon
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - A. Dean Befus
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Marianna Kulka
- National Institute for Nanotechnology, National Research Council, Edmonton, AB, Canada
| |
Collapse
|
44
|
Yamamoto M, Tanaka T, Hozumi Y, Saino-Saito S, Nakano T, Tajima K, Kato T, Goto K. Expression of mRNAs for the diacylglycerol kinase family in immune cells during an inflammatory reaction. Biomed Res 2014; 35:61-8. [PMID: 24573202 DOI: 10.2220/biomedres.35.61] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Phosphoinositide metabolism is intimately involved in cellular signal transduction. In response to extracellular stimuli, it generates diacylglycerol (DG), which serves as a lipid second messenger molecule to activate various proteins in various organs under pathophysiological conditions. Diacylglycerolkinase (DGK) constitutes an enzyme family that catalyzes conversion of DG to phosphatidic acid. It is therefore regarded as a regulator of the DG signal. Previous studies have revealed the critical role of α and ζ types of DGK in T cell functions. Nevertheless, little is known about the expression patterns of the DGK family in immune cells of various kinds. After examination of the expression profile of DGK isozymes in immune cells that are isolated from human blood, we investigated whether their mRNA expression levels would be changed during an inflammatory reaction. Results showed that DGK isozyme mRNAs are widely expressed in immune cells, except for DGKβ and DGKι. During an inflammatory reaction, DGKε mRNA was increased transiently in the initial phase (20-40 min) of stimulation with both LPS and IL-2 in T cell-derived HUT-102 cells and macrophage-derived RAW264 cells. At the organismal level, an intraperitoneal injection of LPS also induced upregulation of DGKε mRNA in the spleen in a similar,but not identical, manner. These results suggest that DGKε is involved in inflammatory processes of the cellular immune system.
Collapse
Affiliation(s)
- Masakazu Yamamoto
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Iida-nishi 2-2-2, Yamagata 990-9585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Melo RCN, Weller PF. Unraveling the complexity of lipid body organelles in human eosinophils. J Leukoc Biol 2014; 96:703-12. [PMID: 25210147 DOI: 10.1189/jlb.3ru0214-110r] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lipid-rich organelles are common in many cell types. In cells, such as adipocytes, these organelles are termed LDs, whereas in other cells, such as leukocytes, they are called LBs. The study of leukocyte LBs has attracted attention as a result of their association with human diseases. In leukocytes, such as eosinophils, LB accumulation has been documented extensively during inflammatory conditions. In these cells, LBs are linked to the regulation of immune responses by compartmentalization of several proteins and lipids involved in the control and biosynthesis of inflammatory mediators (eicosanoids). However, it has been unclear how diverse proteins, including membrane-associated enzymes involved in eicosanoid formation, incorporate into LBs, especially if the internal content of LBs is assumed to consist solely of stores of neutral lipids, as present within adipocyte LDs. Studies of the formation, function, and ultrastructure of LBs in eosinophils have been providing insights pertinent to LBs in other leukocytes. Here, we review current knowledge of the composition and function of leukocyte LBs as provided by studies of human eosinophil LBs, including recognitions of the internal architecture of eosinophil LBs based on 3D electron tomographic analyses.
Collapse
Affiliation(s)
- Rossana C N Melo
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Brazil; and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter F Weller
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Mota LAM, Roberto Neto J, Monteiro VG, Lobato CSS, Oliveira MAD, Cunha MD, D'Ávila H, Seabra SH, Bozza PT, DaMatta RA. Culture of mouse peritoneal macrophages with mouse serum induces lipid bodies that associate with the parasitophorous vacuole and decrease their microbicidal capacity against Toxoplasma gondii. Mem Inst Oswaldo Cruz 2014; 109:767-74. [PMID: 25317704 PMCID: PMC4238769 DOI: 10.1590/0074-0276140119] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 06/26/2014] [Indexed: 11/22/2022] Open
Abstract
Lipid bodies [lipid droplets (LBs)] are lipid-rich organelles involved in lipid metabolism, signalling and inflammation. Recent findings suggest a role for LBs in host response to infection; however, the potential functions of this organelle in Toxoplasma gondii infection and how it alters macrophage microbicidal capacity during infection are not well understood. Here, we investigated the role of host LBs in T. gondii infection in mouse peritoneal macrophages in vitro. Macrophages cultured with mouse serum (MS) had higher numbers of LBs than those cultured in foetal bovine serum and can function as a model to study the role of LBs during intracellular pathogen infection. LBs were found in association with the parasitophorous vacuole, suggesting that T. gondii may benefit from this lipid source. Moreover, increased numbers of macrophage LBs correlated with high prostaglandin E2 (PGE2) production and decreased nitric oxide (NO) synthesis. Accordingly, LB-enriched macrophages cultured with MS were less efficient at controlling T. gondii growth. Treatment of macrophages cultured with MS with indomethacin, an inhibitor of PGE2 production, increased the microbicidal capacity against T. gondii. Collectively, these results suggest that culture with MS caused a decrease in microbicidal activity of macrophages against T. gondii by increasing PGE2 while lowering NO production.
Collapse
Affiliation(s)
- Laura Azeredo Miranda Mota
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - João Roberto Neto
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Verônica Gomes Monteiro
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Caroliny Samary Silva Lobato
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Marco Antonio de Oliveira
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Maura da Cunha
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| | - Heloisa D'Ávila
- Laboratório de Imunofarmacologia, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brasil
| | - Sérgio Henrique Seabra
- Laboratório de Tecnologia em Bioquímica e Microscopia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
| | | | - Renato Augusto DaMatta
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brasil
| |
Collapse
|
47
|
Greineisen WE, Speck M, Shimoda LMN, Sung C, Phan N, Maaetoft-Udsen K, Stokes AJ, Turner H. Lipid body accumulation alters calcium signaling dynamics in immune cells. Cell Calcium 2014; 56:169-80. [PMID: 25016314 DOI: 10.1016/j.ceca.2014.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 10/25/2022]
Abstract
There is well-established variability in the numbers of lipid bodies (LB) in macrophages, eosinophils, and neutrophils. Similarly to the steatosis observed in adipocytes and hepatocytes during hyperinsulinemia and nutrient overload, immune cell LB hyper-accumulate in response to bacterial and parasitic infection and inflammatory presentations. Recently we described that hyperinsulinemia, both in vitro and in vivo, drives steatosis and phenotypic changes in primary and transformed mast cells and basophils. LB reach high numbers in these steatotic cytosols, and here we propose that they could dramatically impact the transcytoplasmic signaling pathways. We compared calcium release and influx responses at the population and single cell level in normal and steatotic model mast cells. At the population level, all aspects of FcɛRI-dependent calcium mobilization, as well as activation of calcium-dependent downstream signaling targets such as NFATC1 phosphorylation are suppressed. At the single cell level, we demonstrate that LB are both sources and sinks of calcium following FcɛRI cross-linking. Unbiased analysis of the impact of the presence of LB on the rate of trans-cytoplasmic calcium signals suggest that LB enrichment accelerates calcium propagation, which may reflect a Bernoulli effect. LB abundance thus impacts this fundamental signaling pathway and its downstream targets.
Collapse
Affiliation(s)
- William E Greineisen
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, HI, United States
| | - Mark Speck
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, HI, United States
| | - Lori M N Shimoda
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, HI, United States
| | - Carl Sung
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, HI, United States
| | - Nolwenn Phan
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, HI, United States
| | - Kristina Maaetoft-Udsen
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, HI, United States
| | - Alexander J Stokes
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, United States
| | - Helen Turner
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, HI, United States; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, United States.
| |
Collapse
|
48
|
Dias FF, Zarantonello VC, Parreira GG, Chiarini-Garcia H, Melo RCN. The intriguing ultrastructure of lipid body organelles within activated macrophages. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2014; 20:869-878. [PMID: 24786359 DOI: 10.1017/s143192761400066x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Macrophages are widely distributed immune system cells with essential functions in tissue homeostasis, apoptotic cell clearance, and first defense in infections. A distinguishing feature of activated macrophages participating in different situations such as inflammatory and metabolic diseases is the presence of increased numbers of lipid-rich organelles, termed lipid bodies (LBs) or lipid droplets, in their cytoplasm. LBs are considered structural markers of activated macrophages and are involved in different functions such as lipid metabolism, intracellular trafficking, and synthesis of inflammatory mediators. In this review, we revisit the distinct morphology of LB organelles actively formed within macrophages in response to infections and cell clearance, taking into account new insights provided by ultrastructural studies. We also discuss the LB interactions within macrophages, revealed by transmission electron microscopy, with a focus on the remarkable LB-phagosome association and discuss potential links between structural aspects and function.
Collapse
Affiliation(s)
- Felipe F Dias
- 1Laboratory of Cellular Biology,Department of Biology,Federal University of Juiz de Fora (UFJF),Juiz de Fora,MG 36036-900,Brazil
| | - Victor C Zarantonello
- 1Laboratory of Cellular Biology,Department of Biology,Federal University of Juiz de Fora (UFJF),Juiz de Fora,MG 36036-900,Brazil
| | - Gleydes G Parreira
- 2Laboratory of Structural Biology and Reproduction,Federal University of Minas Gerais (UFMG),Belo Horizonte,MG 31270-901,Brazil
| | - Hélio Chiarini-Garcia
- 2Laboratory of Structural Biology and Reproduction,Federal University of Minas Gerais (UFMG),Belo Horizonte,MG 31270-901,Brazil
| | - Rossana C N Melo
- 1Laboratory of Cellular Biology,Department of Biology,Federal University of Juiz de Fora (UFJF),Juiz de Fora,MG 36036-900,Brazil
| |
Collapse
|
49
|
Critical role of TLR2 and MyD88 for functional response of macrophages to a group IIA-secreted phospholipase A2 from snake venom. PLoS One 2014; 9:e93741. [PMID: 24718259 PMCID: PMC3981733 DOI: 10.1371/journal.pone.0093741] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/06/2014] [Indexed: 01/10/2023] Open
Abstract
The snake venom MT-III is a group IIA secreted phospholipase A2 (sPLA2) enzyme with functional and structural similarities with mammalian pro-inflammatory sPLA2s of the same group. Previously, we demonstrated that MT-III directly activates the innate inflammatory response of macrophages, including release of inflammatory mediators and formation of lipid droplets (LDs). However, the mechanisms coordinating these processes remain unclear. In the present study, by using TLR2−/− or MyD88−/− or C57BL/6 (WT) male mice, we report that TLR2 and MyD88 signaling have a critical role in MT-III-induced inflammatory response in macrophages. MT-III caused a marked release of PGE2, PGD2, PGJ2, IL-1β and IL-10 and increased the number of LDs in WT macrophages. In MT-III-stimulated TLR2−/− macrophages, formation of LDs and release of eicosanoids and cytokines were abrogated. In MyD88−/− macrophages, MT-III-induced release of PGE2, IL-1β and IL-10 was abrogated, but release of PGD2 and PGJ2 was maintained. In addition, COX-2 protein expression seen in MT-III-stimulated WT macrophages was abolished in both TLR2−/− and MyD88−/− cells, while perilipin 2 expression was abolished only in MyD88−/− cells. We further demonstrated a reduction of saturated, monounsaturated and polyunsaturated fatty acids and a release of the TLR2 agonists palmitic and oleic acid from MT-III-stimulated WT macrophages compared with WT control cells, thus suggesting these fatty acids as major messengers for MT-III-induced engagement of TLR2/MyD88 signaling. Collectively, our findings identify for the first time a TLR2 and MyD88-dependent mechanism that underlies group IIA sPLA2-induced inflammatory response in macrophages.
Collapse
|
50
|
Gomes AF, Magalhães KG, Rodrigues RM, de Carvalho L, Molinaro R, Bozza PT, Barbosa HS. Toxoplasma gondii-skeletal muscle cells interaction increases lipid droplet biogenesis and positively modulates the production of IL-12, IFN-g and PGE2. Parasit Vectors 2014; 7:47. [PMID: 24457118 PMCID: PMC3904159 DOI: 10.1186/1756-3305-7-47] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/21/2014] [Indexed: 12/19/2022] Open
Abstract
Background The interest in the mechanisms involved in Toxoplasma gondii lipid acquisition has steadily increased during the past few decades, but it remains not completely understood. Here, we investigated the biogenesis and the fate of lipid droplets (LD) of skeletal muscle cells (SkMC) during their interaction with T. gondii by confocal and electron microscopy. We also evaluated whether infected SkMC modulates the production of prostaglandin E2 (PGE2), cytokines interleukin-12 (IL-12) and interferon-gamma (INF-g), and also the cyclooxygenase-2 (COX-2) gene induction. Methods Primary culture of skeletal muscle cells were infected with tachyzoites of T. gondii and analysed by confocal microscopy for observation of LD. Ultrastructural cytochemistry was also used for lipid and sarcoplasmatic reticulum (SR) detection. Dosage of cytokines (IL-12 and INF-g) by ELISA technique and enzyme-linked immunoassay (EIA) for PGE2 measurement were employed. The COX-2 gene expression analysis was performed by real time reverse transcriptase polymerase chain reaction (qRT-PCR). Results We demonstrated that T. gondii infection of SkMC leads to increase in LD number and area in a time course dependent manner. Moreover, the ultrastructural analysis demonstrated that SR and LD are in direct contact with parasitophorous vacuole membrane (PVM), within the vacuolar matrix, around it and interacting directly with the membrane of parasite, indicating that LD are recruited and deliver their content inside the parasitophorous vacuole (PV) in T. gondii-infected SkMC. We also observed a positive modulation of the production of IL-12 and IFN-g, increase of COX-2 mRNA levels in the first hour of T. gondii-SkMC interaction and an increase of prostaglandin E2 (PGE2) synthesis from 6 h up to 48 h of infection. Conclusions Taken together, the close association between SR and LD with PV could represent a source of lipids as well as other nutrients for the parasite survival, and together with the increased levels of IL-12, INF-g and inflammatory indicators PGE2 and COX-2 might contribute to the establishment and maintenance of chronic phase of the T. gondii infection in muscle cell.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Helene S Barbosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|